1
|
Liu B, Li Y, Chen H, Li S, Dan X, Xue P, Li Y, Lei L, Fan X. From molecular mechanisms to clinical translation: Silk fibroin-based biomaterials for next-generation wound healing. Int J Biol Macromol 2025; 313:144266. [PMID: 40381758 DOI: 10.1016/j.ijbiomac.2025.144266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Silk fibroin (SF) is a natural polymeric material that has attracted intense research attention in the field of wound healing due to its exceptional mechanical properties, tunable biodegradability, and multifunctional bioactivity. This review systematically summarizes the preparation strategies, functional modifications, and multidimensional application mechanisms of SF and its composite materials in wound healing. The innovative applications of SF in intelligent dressing design, immunometabolic regulation, controlled drug release, stem-cell function modulation, and bioelectrical-activity-mediated microenvironment remodeling is further explored, while analyzing the therapeutic efficacy and cost-effectiveness of SF through clinical translation cases. Distinct from previous reviews, this work not only integrates the latest advances in SF molecular mechanisms and material design but also emphasizes its potential in precision medicine, such as the development of genetically engineered SF for customized immunoregulatory networks. Finally, the article highlights the current challenges in the development of SF materials, including mechanical stability, degradation controllability, and standardization of large-scale production, and envisions future research directions driven by 3D bioprinting and synthetic biology technologies. This review provides a theoretical foundation and technical reference information for the development of efficient, multifunctional, and clinically translatable SF-based materials for application in wound healing.
Collapse
Affiliation(s)
- Bo Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yang Li
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Han Chen
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Songjie Li
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xin Dan
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Ping Xue
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yuli Li
- Shanxi Key Laboratory for Animal Conservation, College of Life Science, Northwest University, Xi'an 710069, China.
| | - Lanjie Lei
- Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China.
| | - Xing Fan
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
2
|
Oh N, Hwang J, Kang MS, Yoo CY, Kwak M, Han DW. Versatile and Marvelous Potentials of Polydeoxyribonucleotide for Tissue Engineering and Regeneration. Biomater Res 2025; 29:0183. [PMID: 40231205 PMCID: PMC11994882 DOI: 10.34133/bmr.0183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 04/16/2025] Open
Abstract
Over the past decade, substantial focus has been placed on polydeoxyribonucleotide (PDRN) due to its promising pharmacological properties, making it a valuable candidate for tissue engineering applications. Accordingly, this paper aims to review and summarize the latest experimental research on PDRN in the context of tissue engineering and regeneration. The unique biochemical mechanisms of PDRN to promote cellular behavior and regeneration are summarized. We categorize commonly utilized PDRN-based tissue engineering fields as neuromuscular tissues, diabetic wound or skin, and bone regeneration. At the same time, we explore scaffold strategies for integrating PDRN into bioceramics, polymers, and cell/tissue-derived materials, along with its combination with photo/electromodulation techniques. Furthermore, we discuss potential opportunities and challenges in translating PDRN-based approaches into clinical practice. We expect future interdisciplinary research and clinical trials to evaluate the long-term efficacy and safety of PDRN while emphasizing standardization and quality control to ensure its consistency and effectiveness in regenerative applications.
Collapse
Affiliation(s)
- Nuri Oh
- Department of Chemistry and Biology,
Korea Science Academy of Korea Advanced Institute of Science and Technology, Busan 47162, Republic of Korea
| | - Juyoung Hwang
- Department of Chemistry,
Pukyong National University, Busan 48513, Republic of Korea
- Smart Gym-Based Translational Research Center for Active Senior’s Healthcare,
Pukyong National University, Busan 48513, Republic of Korea
- Ajou Energy Science Research Center,
Ajou University, Suwon 16499, Republic of Korea
| | - Moon Sung Kang
- Research Institute of Mechanical Technology,
Pusan National University, Busan 46241, Republic of Korea
| | - Chung-Yul Yoo
- Department of Energy Systems Research and Chemistry,
Ajou University, Suwon 16499, Republic of Korea
| | - Minseok Kwak
- Department of Chemistry,
Pukyong National University, Busan 48513, Republic of Korea
- Smart Gym-Based Translational Research Center for Active Senior’s Healthcare,
Pukyong National University, Busan 48513, Republic of Korea
- Industry 4.0 Convergence Bionics Engineering,
Pukyong National University, Busan 48513, Republic of Korea
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering,
Pusan National University, Busan 46241, Republic of Korea
- Institute of Nano-Bio Convergence,
Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
3
|
Shen Z, Zhou X, Zhao M, Li Y. A Structural Optimization Framework for Biodegradable Magnesium Interference Screws. Biomimetics (Basel) 2025; 10:210. [PMID: 40277609 PMCID: PMC12024998 DOI: 10.3390/biomimetics10040210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Biodegradable magnesium alloys have garnered increasing attention in recent years, with magnesium alloy-based biomedical devices being clinically used. Unlike biologically inert metallic materials, magnesium-based medical devices degrade during service, resulting in a mechanical structure that evolves over time. However, there are currently few computer-aided engineering methods specifically tailored for magnesium-based medical devices. This paper introduces a structural optimization framework for Mg-1Ca interference screws, accounting for degradation using a continuum damage model (CDM). The Optimal Latin Hypercube Sampling (OLHS) technique was employed to sample within the design space. Pull-out strengths were used as the optimization objective, which were calculated through finite element analysis (FEA). Both Response Surface Methodology (RSM) and Kriging models were employed as surrogate models and optimized using the Sequential Quadratic Programming (SQP) algorithm. The results from the Kriging model were validated through FEA, and were found to be acceptable. The relationships between the design parameters, the rationale behind the methodology, and its limitations are discussed. Finally, a final design is proposed along with recommendations for interference screw design.
Collapse
Affiliation(s)
- Zhenquan Shen
- Faculty of Artificial Intelligence in Education, Central China Normal University, Wuhan 430079, China
| | - Xiaochen Zhou
- School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Ming Zhao
- Schlumberger Technology Corporation, Houston, TX 77054, USA
| | - Yafei Li
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
4
|
Dunne RA, Dickel DE, Green AM, Kim D, Priddy LB, Priddy MW. Finite Element and Density Functional Theory Modeling Effectively Predict Pitting Degradation of Hydroxyapatite-Coated Pure Magnesium. J Biomed Mater Res B Appl Biomater 2024; 112:e35519. [PMID: 39659047 DOI: 10.1002/jbm.b.35519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 10/11/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024]
Abstract
The emergence of degradable orthopedic implants for fracture fixation may abrogate the need for implant removal surgery and minimize pain associated with permanent implants. Magnesium (Mg) and its alloys are being explored as a biomaterial for degradable implants due to mechanical properties similar to those of bone. Previous in vitro studies have determined the degradation rate of pure Mg to be relatively fast when compared to bone regeneration. Hydroxyapatite (HA), the mineral component of bone, may serve as a surface coating on Mg-based implants to effectively slow and control the degradation rate. The objective of this work was to develop and implement a finite element (FE) model that utilizes a damage evolution law for pitting corrosion to predict the degradation of pure Mg (non-coated) and HA-coated pure Mg (coated) materials simulated in physiological conditions. Finite element analysis (FEA) was performed on a cylindrical Mg specimen (25.4 mm diameter, 8 mm height) through Abaqus/Standard software to incrementally monitor the damage value of each Mg element and subsequently delete fully-degraded elements from the simulation. A Fortran user-material (UMAT) subroutine assigned each element a pitting parameter, controlling the rate of degradation throughout the simulation and providing necessary inputs of elastic material properties and degradation model parameters for pure Mg and HA into Abaqus. The simulations allowed for the visualization of both pure Mg and HA-coated pure Mg degradation over a 120-day period, displaying expected degradation trends such as lower corrosion rates for HA-coated Mg and degradation propagating from the edges inward. Simulation results were calibrated with our prior results from a 30-day experimental degradation study via direct comparison with mass loss over time. Additionally, lower length scale, density functional theory (DFT) simulations were performed to provide physical meaning for the model pitting parameter. The FE simulation was extended to model resin-enclosed pure Mg and HA-coated pure Mg degradation, where only the top surface of the specimen was exposed to the corrosion surface, for investigating changes in Mg surface roughness (height) over time. The impacts of this work include the establishment of a computational model of pure Mg and HA-coated pure Mg degradation calibrated using in vitro degradation data to advance the use of Mg-based biomaterials, and more broadly, to predict degradation rates of next-generation orthopedic implants.
Collapse
Affiliation(s)
- Reese A Dunne
- Michael W. Hall School of Mechanical Engineering, Mississippi State University, Mississippi State, Mississippi, USA
| | - Doyl E Dickel
- Michael W. Hall School of Mechanical Engineering, Mississippi State University, Mississippi State, Mississippi, USA
| | - Addison M Green
- Department of Computer Science and Engineering, Mississippi State University, Mississippi State, Mississippi, USA
| | - Dam Kim
- Michael W. Hall School of Mechanical Engineering, Mississippi State University, Mississippi State, Mississippi, USA
| | - Lauren B Priddy
- Department of Agricultural and Biological Engineering, Mississippi State University, Mississippi State, Mississippi, USA
| | - Matthew W Priddy
- Michael W. Hall School of Mechanical Engineering, Mississippi State University, Mississippi State, Mississippi, USA
| |
Collapse
|
5
|
Bandzerewicz A, Wierzchowski K, Mierzejewska J, Denis P, Gołofit T, Szymczyk-Ziółkowska P, Pilarek M, Gadomska-Gajadhur A. Biological Activity of Poly(1,3-propanediol citrate) Films and Nonwovens: Mechanical, Thermal, Antimicrobial, and Cytotoxicity Studies. Macromol Rapid Commun 2024; 45:e2300452. [PMID: 37838916 DOI: 10.1002/marc.202300452] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/10/2023] [Indexed: 10/16/2023]
Abstract
Polymers are of great interest for medical and cosmeceutical applications. The current trend is to combine materials of natural and synthetic origin in order to obtain products with appropriate mechanical strength and good biocompatibility, additionally biodegradable and bioresorbable. Citric acid, being an important metabolite, is an interesting substance for the synthesis of materials for biomedical applications. Due to the high functionality of the molecule, it is commonly used in biomaterials chemistry as a crosslinking agent. Among citric acid-based biopolyesters, poly(1,8-octanediol citrate) is the best known. It shows application potential in soft tissue engineering. This work focuses on a much less studied polyester, poly(1,3-propanediol citrate). Porous and non-porous materials based on the synthesized polyesters are prepared and characterized, including mechanical, thermal, and surface properties, morphology, and degradation. The main focus is on assessing the biocompatibility and antimicrobial properties of the materials.
Collapse
Affiliation(s)
- Aleksandra Bandzerewicz
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego Street, Warsaw, 00-664, Poland
| | - Kamil Wierzchowski
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1 Street, Warsaw, 00-645, Poland
| | - Jolanta Mierzejewska
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego Street, Warsaw, 00-664, Poland
| | - Piotr Denis
- Laboratory of Polymers and Biomaterials, Institute of Fundamental Technological Research Polish Academy of Sciences, Pawińskiego 5B Street, Warsaw, 02-106, Poland
| | - Tomasz Gołofit
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego Street, Warsaw, 00-664, Poland
| | - Patrycja Szymczyk-Ziółkowska
- Centre for Advanced Manufacturing Technologies-Fraunhofer Project Center, Faculty of Mechanical Engineering, Wroclaw University of Science and Technology, Lukasiewicza 5, Wroclaw, 50-371, Poland
| | - Maciej Pilarek
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1 Street, Warsaw, 00-645, Poland
| | | |
Collapse
|
6
|
Katebifar S, Arul M, Abdulmalik S, Yu X, Alderete JF, Kumbar SG. NOVEL HIGH-STRENGTH POLYESTER COMPOSITE SCAFFOLDS FOR BONE REGENERATION. POLYM ADVAN TECHNOL 2023; 34:3770-3791. [PMID: 38312483 PMCID: PMC10836609 DOI: 10.1002/pat.6178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/14/2023] [Indexed: 02/06/2024]
Abstract
Repair of critical sized bone defects, particularly in load-bearing areas, is a major clinical problem that requires surgical intervention and implantation of biological or engineered grafts. For load-bearing sites, it is essential to use engineered grafts that have both sufficient mechanical strength and appropriate pore properties to support bone repair and tissue regeneration. Unfortunately, the mechanical properties of such grafts are often compromised due to the creation of pores required to facilitate tissue ingrowth following implantation. To overcome the limitations associated with porous scaffolds and their reduced mechanical strength, we have developed a methodology for creating a solid structure that retains its bulk mechanical properties while also evolving into a porous structure in a biological environment through degradation and erosion. In this study, we utilized polyesters that have been approved by the FDA, including poly (lactic acid) (PLA), poly(glycolic acid) (PGA), their copolymer PLGA (PLGA, with a ratio of 85:15 and 50:50 of PLA:PGA), and poly(caprolactone) (PCL). These polymers and their ceramic composites with tricalcium phosphate (TCP) were compression molded into solid forms, which exhibited mechanical properties with compressive modulus as high as 2745 ± 364 MPa within the range of human trabecular bone and in the lower range of human cortical bone. The use of fast-degrading PLGA (50:50) and PGA as porogens allowed the formation of pores within the solid structures due to their degradation, and the TCP acts as a buffering agent to neutralize their acidic degradation byproducts. These scaffolds facilitated the growth of new blood vessels and tissue ingrowth in a subcutaneous implantation model. In addition, in a rat critical-sized mandibular bone defects these scaffolds supported bone growth with 70% of new bone volume fraction. Furthermore, the extent of bone regeneration was found to be higher for the scaffolds with bone morphogenic proteins (BMP2), indicating their suitability for bone repair and regeneration.
Collapse
Affiliation(s)
- Sara Katebifar
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Michael Arul
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Sama Abdulmalik
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Xiaojun Yu
- Department of Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Joseph F. Alderete
- Departments of Orthopedic Surgery, Brooke Army Medical Center, Joint Base San Antonio, Texas
| | - Sangamesh G. Kumbar
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, 06269, USA
| |
Collapse
|
7
|
Liang NE, Griffin MF, Berry CE, Parker JB, Downer MA, Wan DC, Longaker MT. Attenuating Chronic Fibrosis: Decreasing Foreign Body Response with Acellular Dermal Matrix. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:671-680. [PMID: 37212342 DOI: 10.1089/ten.teb.2023.0060] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Surgical implants are increasingly used across multiple medical disciplines, with applications ranging from tissue reconstruction to improving compromised organ and limb function. Despite their significant potential for improving health and quality of life, biomaterial implant function is severely limited by the body's immune response to its presence: this is known as the foreign body response (FBR) and is characterized by chronic inflammation and fibrotic capsule formation. This response can result in life-threatening sequelae such as implant malfunction, superimposed infection, and associated vessel thrombosis, in addition to soft tissue disfigurement. Patients may require frequent medical visits, as well as repeated invasive procedures, increasing the burden on an already strained health care system. Currently, the FBR and the cells and molecular mechanisms that mediate it are poorly understood. With applications across a wide array of surgical specialties, acellular dermal matrix (ADM) has emerged as a potential solution to the fibrotic reaction seen with FBR. Although the mechanisms by which ADM decreases chronic fibrosis remain to be clearly characterized, animal studies across diverse surgical models point to its biomimetic properties that facilitate decreased periprosthetic inflammation and improved host cell incorporation. Impact Statement Foreign body response (FBR) is a significant limitation to the use of implantable biomaterials. Acellular dermal matrix (ADM) has been observed to decrease the fibrotic reaction seen with FBR, although its mechanistic details are poorly understood. This review is dedicated to summarizing the primary literature on the biology of FBR in the context of ADM use, using surgical models in breast reconstruction, abdominal and chest wall repair, and pelvic reconstruction. This article will provide readers with an overarching review of shared mechanisms for ADM across multiple surgical models and diverse anatomical applications.
Collapse
Affiliation(s)
- Norah E Liang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michelle F Griffin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Charlotte E Berry
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jennifer B Parker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Mauricio A Downer
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Derrick C Wan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael T Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
8
|
Kurowiak J, Klekiel T, Będziński R. Biodegradable Polymers in Biomedical Applications: A Review-Developments, Perspectives and Future Challenges. Int J Mol Sci 2023; 24:16952. [PMID: 38069272 PMCID: PMC10707259 DOI: 10.3390/ijms242316952] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Biodegradable polymers are materials that, thanks to their remarkable properties, are widely understood to be suitable for use in scientific fields such as tissue engineering and materials engineering. Due to the alarming increase in the number of diagnosed diseases and conditions, polymers are of great interest in biomedical applications especially. The use of biodegradable polymers in biomedicine is constantly expanding. The application of new techniques or the improvement of existing ones makes it possible to produce materials with desired properties, such as mechanical strength, controlled degradation time and rate and antibacterial and antimicrobial properties. In addition, these materials can take virtually unlimited shapes as a result of appropriate design. This is additionally desirable when it is necessary to develop new structures that support or restore the proper functioning of systems in the body.
Collapse
Affiliation(s)
| | | | - Romuald Będziński
- Department of Biomedical Engineering, Institute of Material and Biomedical Engineering, Faculty of Mechanical Engineering, University of Zielona Góra, Licealna 9 Street, 65-417 Zielona Gora, Poland; (J.K.); (T.K.)
| |
Collapse
|
9
|
Kim K, Su Y, Kucine AJ, Cheng K, Zhu D. Guided Bone Regeneration Using Barrier Membrane in Dental Applications. ACS Biomater Sci Eng 2023; 9:5457-5478. [PMID: 37650638 DOI: 10.1021/acsbiomaterials.3c00690] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Guided bone regeneration (GBR) is a widely used technique in preclinical and clinical studies due to its predictability. Its main purpose is to prevent the migration of soft tissue into the osseous wound space, while allowing osseous cells to migrate to the site. GBR is classified into two main categories: resorbable and non-resorbable membranes. Resorbable membranes do not require a second surgery but tend to have a short resorption period. Conversely, non-resorbable membranes maintain their mechanical strength and prevent collapse. However, they require removal and are susceptible to membrane exposure. GBR is often used with bone substitute graft materials to fill the defect space and protect the bone graft. The membrane can also undergo various modifications, such as surface modification and biological factor loading, to improve barrier functions and bone regeneration. In addition, bone regeneration is largely related to osteoimmunology, a new field that focuses on the interactions between bone and the immune system. Understanding these interactions can help in developing new treatments for bone diseases and injuries. Overall, GBR has the potential to be a powerful tool in promoting bone regeneration. Further research in this area could lead to advancements in the field of bone healing. This review will highlight resorbable and non-resorbable membranes with cellular responses during bone regeneration, provide insights into immunological response during bone remodeling, and discuss antibacterial features.
Collapse
Affiliation(s)
- Kakyung Kim
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Yingchao Su
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Allan J Kucine
- Department of Oral and Maxillofacial Surgery, Stony Brook University, Stony Brook, New York 11794, United States
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Donghui Zhu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
10
|
Patel SK, Khan S, Dasari V, Gupta S. Beyond Pain Relief: An In-Depth Review of Vertebral Height Restoration After Balloon Kyphoplasty in Vertebral Compression Fractures. Cureus 2023; 15:e46124. [PMID: 37900521 PMCID: PMC10612383 DOI: 10.7759/cureus.46124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023] Open
Abstract
This comprehensive review delves into the intricate landscape of vertebral height restoration after balloon kyphoplasty in cases of vertebral compression fractures. With a comprehensive examination of procedural intricacies, radiological evaluations, clinical outcomes, and influential factors, a nuanced comprehension unfolds. Beyond its immediate alleviation of pain, vertebral height restoration emerges as a linchpin in enhancing spinal alignment, fostering functional recuperation, and augmenting the overall quality of life. This review underscores the pivotal role of balloon kyphoplasty, transcending its mere medical utility to become a conduit for renewed independence and well-being among individuals grappling with vertebral compression fractures. The ongoing advancements in medical science and the continued pursuit of research stand poised to amplify the significance of vertebral height restoration, manifesting a promising horizon for individuals seeking respite from pain, a revitalised capacity for movement, and a life unburdened by its constraints.
Collapse
Affiliation(s)
- Siddharth K Patel
- Orthopaedics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sohael Khan
- Orthopaedics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ventaktesh Dasari
- Orthopaedics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Suvarn Gupta
- Orthopaedics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
11
|
Jiang EY, Desroches ST, Mikos AG. Particle carriers for controlled release of peptides. J Control Release 2023; 360:953-968. [PMID: 37004797 DOI: 10.1016/j.jconrel.2023.03.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/28/2023] [Indexed: 04/04/2023]
Abstract
There has been growing discovery and use of therapeutic peptides in drug delivery and tissue engineering. Peptides are smaller than proteins and can be formulated into drug delivery systems without significant loss of their bioactivity, which remains a concern with proteins. However, the smaller size of peptides has made the controlled release of these bioactive molecules from carriers challenging. Thus, there has been increasing development of carriers to improve the controlled release of peptides by leveraging hydrophobic and electrostatic interactions between the peptide and the carrier. The focus of this review paper is to critically discuss synthetic and natural nanoparticles and microparticles that have been investigated for the controlled delivery of peptides with emphasis on the underlying interactions.
Collapse
Affiliation(s)
- Emily Y Jiang
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, USA
| | - Shelby T Desroches
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, USA
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Lee H, Shin DY, Na Y, Han G, Kim J, Kim N, Bang SJ, Kang HS, Oh S, Yoon CB, Park J, Kim HE, Jung HD, Kang MH. Antibacterial PLA/Mg composite with enhanced mechanical and biological performance for biodegradable orthopedic implants. BIOMATERIALS ADVANCES 2023; 152:213523. [PMID: 37336010 DOI: 10.1016/j.bioadv.2023.213523] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/29/2023] [Accepted: 06/12/2023] [Indexed: 06/21/2023]
Abstract
Biodegradability, bone-healing rate, and prevention of bacterial infection are critical factors for orthopedic implants. Polylactic acid (PLA) is a good candidate biodegradable material; however, it has insufficient mechanical strength and bioactivity for orthopedic implants. Magnesium (Mg), has good bioactivity, biodegradability, and sufficient mechanical properties, similar to that of bone. Moreover, Mg has an inherent antibacterial property via a photothermal effect, which generates localized heat, thus preventing bacterial infection. Therefore, Mg is a good candidate material for PLA composites, to improve their mechanical and biological performance and add an antibacterial property. Herein, we fabricated an antibacterial PLA/Mg composite for enhanced mechanical and biological performance with an antibacterial property for application as biodegradable orthopedic implants. The composite was fabricated with 15 and 30 vol% of Mg homogeneously dispersed in PLA without the generation of a defect using a high-shear mixer. The composites exhibited an enhanced compressive strength of 107.3 and 93.2 MPa, and stiffness of 2.3 and 2.5 GPa, respectively, compared with those of pure PLA which were 68.8 MPa and 1.6 GPa, respectively. Moreover, the PLA/Mg composite at 15 vol% Mg exhibited significant improvement of biological performance in terms of enhanced initial cell attachment and cell proliferation, whereas the composite at 30 vol% Mg showed deteriorated cell proliferation and differentiation because of the rapid degradation of the Mg particles. In turn, the PLA/Mg composites exerted an antibacterial effect based on the inherent antibacterial property of Mg as well as the photothermal effect induced by near-infrared (NIR) treatment, which can minimize infection after implantation surgery. Therefore, antibacterial PLA/Mg composites with enhanced mechanical and biological performance may be a candidate material with great potential for biodegradable orthopedic implants.
Collapse
Affiliation(s)
- Hyun Lee
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Da-Young Shin
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Yuhyun Na
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Ginam Han
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Joodeok Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Nahyun Kim
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Seo-Jun Bang
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Hyeong Seok Kang
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - SeKwon Oh
- Research Institute of Advanced Manufacturing & Materials Technology, Korea Institute of Industrial Technology, Incheon 21999, Republic of Korea
| | - Chang-Bun Yoon
- Department of Advanced Materials Engineering, Tech University of Korea, Siheung-si 15073, Republic of Korea
| | - Jungwon Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea; Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul 08826, Republic of Korea; Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea; Advanced Institutes of Convergence Technology, Seoul National University, Suwon-si 16229, Republic of Korea
| | - Hyoun-Ee Kim
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyun-Do Jung
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Min-Ho Kang
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea; Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea.
| |
Collapse
|
13
|
Sun L, Sun X, Ruan W, Che G, Zhu F, Liu C, Wan M. Mechanism of remodeling and local effects in vivo of a new injectable cosmetic filler. Sci Rep 2023; 13:9599. [PMID: 37311816 DOI: 10.1038/s41598-023-36510-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023] Open
Abstract
By studying the local effects of a new type of injectable cosmetic filler implanted into the animal to explore the mechanism of remodeling and cosmetic effect of this kind of product. Take 12 rabbits and select 4 implantation points on both sides of the spine, respectively, and implant the test sample (PLLA) and negative control sample (HDPE) into the subcutaneous tissues on both sides. In the same way, take another 12 rabbits and implant the marketing control sample (cross-linked sodium hyaluronate) and negative control sample (HDPE) into the subcutaneous tissues on both sides. The animals were executed at 1 week, 4 weeks, 13 weeks and 52 weeks respectively, and Hematoxylin-Eosin staining, Masson trichrome staining and immunofluorescence staining were performed to characterize the local effects in vivo and the expression of type I collagen (Col. I), type III collagen (Col.III) and matrix metalloproteinase 9 (MMP-9). Good histocompatibility of the test sample and the marketing control sample were found. The foreign body reaction of marketing control sample was more intense than that of the test sample after 13 weeks. The foreign body reaction of testing sample was more intense after 52 weeks, while that of the marketing control sample was more stable. With the process of tissue repair, the collagen fibers of test samples and marketing control samples gradually increased after implantation. Type I collagen was mainly found inside the fiber capsule, while type III collagen was mainly found outside. The positive expression of matrix metalloproteinase 9 gradually increased, the positive expression of test samples increased significantly after 52 weeks, while that of marketing control samples did not change significantly. Good histocompatibility of PLLA filler is found. Matrix metalloproteinase 9 participates in foreign body reaction and collagen formation, which can reflect the process of tissue remodeling.
Collapse
Affiliation(s)
- Likui Sun
- Shandong Institute of Medical Device and Pharmaceutical Packaging Inspection (National Medical Products Administration Jinan Quality Supervision and Inspection Center for Medical Devices), which is NMPA Key laboratory for Safety Evaluation of Biomaterials and Medical Devices, Key lab of biological evaluation of medical devices of Shandong province, Jinan, 250101, China.
| | - Xiaoxia Sun
- Shandong Institute of Medical Device and Pharmaceutical Packaging Inspection (National Medical Products Administration Jinan Quality Supervision and Inspection Center for Medical Devices), which is NMPA Key laboratory for Safety Evaluation of Biomaterials and Medical Devices, Key lab of biological evaluation of medical devices of Shandong province, Jinan, 250101, China
| | - Wenting Ruan
- Shandong Institute of Medical Device and Pharmaceutical Packaging Inspection (National Medical Products Administration Jinan Quality Supervision and Inspection Center for Medical Devices), which is NMPA Key laboratory for Safety Evaluation of Biomaterials and Medical Devices, Key lab of biological evaluation of medical devices of Shandong province, Jinan, 250101, China
| | - Guoxi Che
- Shandong Institute of Medical Device and Pharmaceutical Packaging Inspection (National Medical Products Administration Jinan Quality Supervision and Inspection Center for Medical Devices), which is NMPA Key laboratory for Safety Evaluation of Biomaterials and Medical Devices, Key lab of biological evaluation of medical devices of Shandong province, Jinan, 250101, China
| | - Fuyu Zhu
- Shandong Institute of Medical Device and Pharmaceutical Packaging Inspection (National Medical Products Administration Jinan Quality Supervision and Inspection Center for Medical Devices), which is NMPA Key laboratory for Safety Evaluation of Biomaterials and Medical Devices, Key lab of biological evaluation of medical devices of Shandong province, Jinan, 250101, China
| | - Chenghu Liu
- Shandong Institute of Medical Device and Pharmaceutical Packaging Inspection (National Medical Products Administration Jinan Quality Supervision and Inspection Center for Medical Devices), which is NMPA Key laboratory for Safety Evaluation of Biomaterials and Medical Devices, Key lab of biological evaluation of medical devices of Shandong province, Jinan, 250101, China
| | - Min Wan
- Shandong Institute of Medical Device and Pharmaceutical Packaging Inspection (National Medical Products Administration Jinan Quality Supervision and Inspection Center for Medical Devices), which is NMPA Key laboratory for Safety Evaluation of Biomaterials and Medical Devices, Key lab of biological evaluation of medical devices of Shandong province, Jinan, 250101, China
| |
Collapse
|
14
|
Kim HS, Kumbar SG, Nukavarapu SP. Amorphous silica fiber matrix biomaterials: An analysis of material synthesis and characterization for tissue engineering. Bioact Mater 2023; 19:155-166. [PMID: 35441118 PMCID: PMC9006749 DOI: 10.1016/j.bioactmat.2022.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/09/2022] [Accepted: 04/02/2022] [Indexed: 11/30/2022] Open
Abstract
Silica biomaterials including Bioglass offer great biocompatibility and bioactivity but fail to provide pore and degradation features needed for tissue engineering. Herein we report on the synthesis and characterization of novel amorphous silica fiber matrices to overcome these limitations. Amorphous silica fibers were fused by sintering to produce porous matrices. The effects of sacrificial polymer additives such as polyvinyl alcohol (PVA) and cellulose fibers (CF) on the sintering process were also studied. The resulting matrices formed between sintering temperatures of 1,350–1,550 °C retained their fiber structures. The matrices presented pores in the range of 50–200 μm while higher sintering temperatures resulted in increased pore diameter. PVA addition to silica significantly reduced the pore diameter and porosity compared with silica matrices with or without the addition of CF. The PVA additive morphologically appeared to fuse the silica fibers to a greater extent and resulted in significantly higher compressive modulus and strength than the rest of the matrices synthesized. These matrices lost roughly 30% of their original mass in an in vitro degradation study over 40 weeks. All matrices absorbed 500 wt% of water and did not change in their overall morphology, size, or shape with hydration. These fiber matrices supported human mesenchymal stem cell adhesion, proliferation, and mineralized matrix production. Amorphous silica fiber biomaterials/matrices reported here are biodegradable and porous and closely resemble the native extracellular matrix structure and water absorption capacity. Extending the methodology reported here to alter matrix properties may lead to a variety of tissue engineering, implant, and drug delivery applications.
Collapse
Affiliation(s)
- Hyun S. Kim
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Sangamesh G. Kumbar
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Material Science and Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Syam P. Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Material Science and Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA
- Corresponding author. Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
15
|
Brito J, Andrianov AK, Sukhishvili SA. Factors Controlling Degradation of Biologically Relevant Synthetic Polymers in Solution and Solid State. ACS APPLIED BIO MATERIALS 2022; 5:5057-5076. [PMID: 36206552 DOI: 10.1021/acsabm.2c00694] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The field of biodegradable synthetic polymers, which is central for regenerative engineering and drug delivery applications, encompasses a multitude of hydrolytically sensitive macromolecular structures and diverse processing approaches. The ideal degradation behavior for a specific life science application must comply with a set of requirements, which include a clinically relevant kinetic profile, adequate biocompatibility, benign degradation products, and controlled structural evolution. Although significant advances have been made in tailoring materials characteristics to satisfy these requirements, the impacts of autocatalytic reactions and microenvironments are often overlooked resulting in uncontrollable and unpredictable outcomes. Therefore, roles of surface versus bulk erosion, in situ microenvironment, and autocatalytic mechanisms should be understood to enable rational design of degradable systems. In an attempt to individually evaluate the physical state and form factors influencing autocatalytic hydrolysis of degradable polymers, this Review follows a hierarchical analysis that starts with hydrolytic degradation of water-soluble polymers before building up to 2D-like materials, such as ultrathin coatings and capsules, and then to solid-state degradation. We argue that chemical reactivity largely governs solution degradation while diffusivity and geometry control the degradation of bulk materials, with thin "2D" materials remaining largely unexplored. Following this classification, this Review explores techniques to analyze degradation in vitro and in vivo and summarizes recent advances toward understanding degradation behavior for traditional and innovative polymer systems. Finally, we highlight challenges encountered in analytical methodology and standardization of results and provide perspective on the future trends in the development of biodegradable polymers.
Collapse
Affiliation(s)
- Jordan Brito
- Department of Materials Science & Engineering, Texas A&M University, College Station, Texas77843, United States
| | - Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland20850, United States
| | - Svetlana A Sukhishvili
- Department of Materials Science & Engineering, Texas A&M University, College Station, Texas77843, United States
| |
Collapse
|
16
|
Gareb B, van Bakelen NB, Driessen L, Buma P, Kuipers J, Grijpma DW, Vissink A, Bos RR, van Minnen B. Biocompatibility and degradation comparisons of four biodegradable copolymeric osteosynthesis systems used in maxillofacial surgery: A goat model with four years follow-up. Bioact Mater 2022; 17:439-456. [PMID: 35386449 PMCID: PMC8961280 DOI: 10.1016/j.bioactmat.2022.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/10/2021] [Accepted: 01/08/2022] [Indexed: 01/15/2023] Open
Abstract
Applying biodegradable osteosyntheses avoids the disadvantages of titanium osteosyntheses. However, foreign-body reactions remain a major concern and evidence of complete resorption is lacking. This study compared the physico-chemical properties, histological response and radiographs of four copolymeric biodegradable osteosynthesis systems in a goat model with 48-months follow-up. The systems were implanted subperiosteally in both tibia and radius of 12 Dutch White goats. The BioSorb FX [poly(70LLA-co-30DLLA)], Inion CPS [poly([70–78.5]LLA-co-[16–24]DLLA-co-4TMC)], SonicWeld Rx [poly(DLLA)], LactoSorb [poly(82LLA-co-18GA)] systems and a negative control were randomly implanted in each extremity. Samples were assessed at 6-, 12-, 18-, 24-, 36-, and 48-month follow-up. Surface topography was performed using scanning electron microscopy (SEM). Differential scanning calorimetry and gel permeation chromatography were performed on initial and explanted samples. Histological sections were systematically assessed by two blinded researchers using (polarized) light microscopy, SEM and energy-dispersive X-ray analysis. The SonicWeld Rx system was amorphous while the others were semi-crystalline. Foreign-body reactions were not observed during the complete follow-up. The SonicWeld Rx and LactoSorb systems reached bone percentages of negative controls after 18 months while the BioSorb Fx and Inion CPS systems reached these levels after 36 months. The SonicWeld Rx system showed the most predictable degradation profile. All the biodegradable systems were safe to use and well-tolerated (i.e., complete implant replacement by bone, no clinical or histological foreign body reactions, no [sterile] abscess formation, no re-interventions needed), but nanoscale residual polymeric fragments were observed at every system's assessment. Foreign-body reactions are a major concern of biodegradable osteosyntheses. Amorphous poly(DLLA) showed the most predictable degradation profile. Nanoscale residual polymeric fragments could still be observed after 4 years.
Collapse
|
17
|
Lozhkomoev AS, Buyakov AS, Kazantsev SO, Senkina EI, Krinitcyn MG, Ivanyuk VA, Sharipova AF, Lerner MI. Preparation and Properties of Iron Nanoparticle-Based Macroporous Scaffolds for Biodegradable Implants. MATERIALS (BASEL, SWITZERLAND) 2022; 15:4900. [PMID: 35888367 PMCID: PMC9317871 DOI: 10.3390/ma15144900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 06/29/2022] [Accepted: 07/12/2022] [Indexed: 02/01/2023]
Abstract
Fe-based scaffolds are of particular interest in the technology of biodegradable implants due to their high mechanical properties and biocompatibility. In the present work, using an electroexplosive Fe nanopowder and NaCl particles 100-200 µm in size as a porogen, scaffolds with a porosity of about 70 ± 0.8% were obtained. The effect of the sintering temperature on the structure, composition, and mechanical characteristics of the scaffolds was considered. The optimum parameters of the sintering process were determined, allowing us to obtain samples characterized by plastic deformation and a yield strength of up to 16.2 MPa. The degradation of the scaffolds sintered at 1000 and 1100 °C in 0.9 wt.% NaCl solution for 28 days resulted in a decrease in their strength by 23% and 17%, respectively.
Collapse
Affiliation(s)
- Aleksandr S. Lozhkomoev
- Institute of Strength Physics and Materials Science of the Siberian Branch of the Russian Academy of Sciences (ISPMS SB RAS), 634021 Tomsk, Russia; (A.S.B.); (S.O.K.); (E.I.S.); (M.G.K.); (M.I.L.)
| | - Ales S. Buyakov
- Institute of Strength Physics and Materials Science of the Siberian Branch of the Russian Academy of Sciences (ISPMS SB RAS), 634021 Tomsk, Russia; (A.S.B.); (S.O.K.); (E.I.S.); (M.G.K.); (M.I.L.)
| | - Sergey O. Kazantsev
- Institute of Strength Physics and Materials Science of the Siberian Branch of the Russian Academy of Sciences (ISPMS SB RAS), 634021 Tomsk, Russia; (A.S.B.); (S.O.K.); (E.I.S.); (M.G.K.); (M.I.L.)
| | - Elena I. Senkina
- Institute of Strength Physics and Materials Science of the Siberian Branch of the Russian Academy of Sciences (ISPMS SB RAS), 634021 Tomsk, Russia; (A.S.B.); (S.O.K.); (E.I.S.); (M.G.K.); (M.I.L.)
| | - Maksim G. Krinitcyn
- Institute of Strength Physics and Materials Science of the Siberian Branch of the Russian Academy of Sciences (ISPMS SB RAS), 634021 Tomsk, Russia; (A.S.B.); (S.O.K.); (E.I.S.); (M.G.K.); (M.I.L.)
| | - Valeria A. Ivanyuk
- Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia;
| | - Aliya F. Sharipova
- Department of Materials Science and Engineering, Technion, Haifa 3200003, Israel;
| | - Marat I. Lerner
- Institute of Strength Physics and Materials Science of the Siberian Branch of the Russian Academy of Sciences (ISPMS SB RAS), 634021 Tomsk, Russia; (A.S.B.); (S.O.K.); (E.I.S.); (M.G.K.); (M.I.L.)
| |
Collapse
|
18
|
Titanium or Biodegradable Osteosynthesis in Maxillofacial Surgery? In Vitro and In Vivo Performances. Polymers (Basel) 2022; 14:polym14142782. [PMID: 35890557 PMCID: PMC9316877 DOI: 10.3390/polym14142782] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/30/2022] [Accepted: 07/02/2022] [Indexed: 02/06/2023] Open
Abstract
Osteosynthesis systems are used to fixate bone segments in maxillofacial surgery. Titanium osteosynthesis systems are currently the gold standard. However, the disadvantages result in symptomatic removal in up to 40% of cases. Biodegradable osteosynthesis systems, composed of degradable polymers, could reduce the need for removal of osteosynthesis systems while avoiding the aforementioned disadvantages of titanium osteosyntheses. However, disadvantages of biodegradable systems include decreased mechanical properties and possible foreign body reactions. In this review, the literature that focused on the in vitro and in vivo performances of biodegradable and titanium osteosyntheses is discussed. The focus was on factors underlying the favorable clinical outcome of osteosyntheses, including the degradation characteristics of biodegradable osteosyntheses and the host response they elicit. Furthermore, recommendations for clinical usage and future research are given. Based on the available (clinical) evidence, biodegradable copolymeric osteosyntheses are a viable alternative to titanium osteosyntheses when applied to treat maxillofacial trauma, with similar efficacy and significantly lower symptomatic osteosynthesis removal. For orthognathic surgery, biodegradable copolymeric osteosyntheses are a valid alternative to titanium osteosyntheses, but a longer operation time is needed. An osteosynthesis system composed of an amorphous copolymer, preferably using ultrasound welding with well-contoured shapes and sufficient mechanical properties, has the greatest potential as a biocompatible biodegradable copolymeric osteosynthesis system. Future research should focus on surface modifications (e.g., nanogel coatings) and novel biodegradable materials (e.g., magnesium alloys and silk) to address the disadvantages of current osteosynthesis systems.
Collapse
|
19
|
A Simple Replica Method as the Way to Obtain a Morphologically and Mechanically Bone-like Iron-Based Biodegradable Material. MATERIALS 2022; 15:ma15134552. [PMID: 35806677 PMCID: PMC9267498 DOI: 10.3390/ma15134552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/04/2022]
Abstract
Porous iron-based scaffolds were prepared by the simple replica method using polyurethane foam as a template and applying the sintering process in a tube furnace. Their surface morphology was characterized using scanning electron microscopy (SEM) and phase homogeneity was confirmed using X-ray diffraction (XRD). Corrosion behavior was determined using immersion and potentiodynamic polarization methods in phosphate buffered saline (PBS). The surface energy was calculated by studying the changes of enthalpy of calorimetric immersion. A preliminary biological test was also carried out and was done using the albumin adsorption procedure. Results of our work showed that in using the simple replica method it is possible to obtain iron biomaterial with morphology and mechanical properties almost identical to bones, and possessing adequate wettability, which gives the potential to use this material as biomaterial for scaffolds in orthopedics.
Collapse
|
20
|
ABSTRACTS (BY NUMBER). Tissue Eng Part A 2022. [DOI: 10.1089/ten.tea.2022.29025.abstracts] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
21
|
Chen D, Xi Y, Zhang S, Weng L, Dong Z, Chen C, Wu T, Xiao J. Curcumin attenuates inflammation of Macrophage-derived foam cells treated with Poly-L-lactic acid degradation via PPARγ signaling pathway. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2022; 33:33. [PMID: 35303193 PMCID: PMC8933344 DOI: 10.1007/s10856-022-06654-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/31/2022] [Indexed: 05/12/2023]
Abstract
Poly-L-lactic acid (PLLA) is considered to be a promising candidate material for biodegradable vascular scaffolds (BVS) in percutaneous coronary intervention (PCI). But, PLLA-BVS also faces the challenge of thrombosis (ST) and in-stent restenosis (ISR) caused by in-stent neo-atherosclerosis (ISNA) associated with inflammatory reactions in macrophage-derived foam cells. Our previous studies have confirmed that curcumin alleviates PLLA-induced injury and inflammation in vascular endothelial cells, but it remains unclear whether curcumin can alleviate the effect of inflammatory reactions in macrophage-derived foam cells while treated with degraded product of PLLA. In this study, PLLA-BVS was implanted in the porcine coronary artery to examine increased macrophages and inflammatory cytokines such as NF-κb and TNF-α by histology and immunohistochemistry. In vitro, macrophage-derived foam cells were induced by Ox-LDL and observed by Oil Red Staining. Foam cells were treated with pre-degraded PLLA powder, curcumin and PPARγ inhibitor GW9662, and the expression of IL-6, IL-10, TNF-α, NF-κb, PLA2 and PPARγ were investigated by ELISA or RT-qPCR. This study demonstrated that the macrophages and inflammatory factors increased after PLLA-BVS implantation in vivo, and foam cells derived from macrophages promoted inflammation by products of PLLA degradation in vitro. This present study was found that the inflammation of foam cells at the microenvironment of PLLA degraded products were significantly increased, and curcumin can attenuate the inflammation caused by the PLLA degradation via PPARγ signal pathway. In addition, curcumin should be further studied experimentally in vivo experiments on animal models as a potential therapeutic to reduce ISNA of PLLA-BVS. Graphical abstract.
Collapse
Affiliation(s)
- Dongping Chen
- Central Laboratory, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Yangbo Xi
- Department of Cardiology, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
- Department of The First Clinical Medical College, Jinan University, Guangzhou, China
| | - Suzhen Zhang
- Central Laboratory, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Linsheng Weng
- Department of Cardiology, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
- Intensive Care Unit of Foshan Women and Children Hospital, Foshan, China
| | - Zhihui Dong
- Central Laboratory, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Can Chen
- Department of pathology, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Tim Wu
- Dongguan TT Medical,Inc, Dongguan, China
- VasoTech, Inc., Lowell, MA, USA
| | - Jianmin Xiao
- Central Laboratory, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China.
- Department of Cardiology, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China.
- Department of The First Clinical Medical College, Jinan University, Guangzhou, China.
| |
Collapse
|
22
|
Kim DS, Lee JK, Kim JH, Lee J, Kim DS, An S, Park SB, Kim TH, Rim JS, Lee S, Han DK. Advanced PLGA hybrid scaffold with a bioactive PDRN/BMP2 nanocomplex for angiogenesis and bone regeneration using human fetal MSCs. SCIENCE ADVANCES 2021; 7:eabj1083. [PMID: 34878837 PMCID: PMC8654289 DOI: 10.1126/sciadv.abj1083] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/16/2021] [Indexed: 05/14/2023]
Abstract
Biodegradable polymers have been used with various systems for tissue engineering. Among them, poly(lactic-co-glycolic) acid (PLGA) has been widely used as a biomaterial for bone regeneration because of its great biocompatibility and biodegradability properties. However, there remain substantial cruxes that the by-products of PLGA result in an acidic environment at the implanting site, and the polymer has a weak mechanical property. In our previous study, magnesium hydroxide (MH) and bone extracellular matrix are combined with a PLGA scaffold (PME) to improve anti-inflammation and mechanical properties and osteoconductivity. In the present study, the development of a bioactive nanocomplex (NC) formed along with polydeoxyribonucleotide and bone morphogenetic protein 2 (BMP2) provides synergistic abilities in angiogenesis and bone regeneration. This PME hybrid scaffold immobilized with NC (PME/NC) achieves outstanding performance in anti-inflammation, angiogenesis, and osteogenesis. Such an advanced PME/NC scaffold suggests an integrated bone graft substitute for bone regeneration.
Collapse
Affiliation(s)
- Da-Seul Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Jun Hyuk Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Jaemin Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Dong Seon Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Sanghyun An
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Sung-Bin Park
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jong Seop Rim
- Fetal Stem Cell Research Center, CHA Advanced Research Institute, Gyeonggi-do 13488, Republic of Korea
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Gyeonggi-do 13496, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| |
Collapse
|
23
|
Prabhath A, Vernekar VN, Vasu V, Badon M, Avochinou JE, Asandei AD, Kumbar SG, Weber E, Laurencin CT. Kinetic degradation and biocompatibility evaluation of polycaprolactone-based biologics delivery matrices for regenerative engineering of the rotator cuff. J Biomed Mater Res A 2021; 109:2137-2153. [PMID: 33974735 PMCID: PMC8440380 DOI: 10.1002/jbm.a.37200] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 11/06/2022]
Abstract
Whereas synthetic biodegradable polymers have been successfully applied for the delivery of biologics in other tissues, the anatomical complexity, poor blood supply, and reduced clearance of degradation byproducts in the rotator cuff create unique design challenges for implantable biomaterials. Here, we investigated lower molecular weight poly-lactic acid co-epsilon-caprolactone (PLA-CL) formulations with varying molecular weight and film casting concentrations as potential matrices for the therapeutic delivery of biologics in the rotator cuff. Matrices were fabricated with target footprint dimensions to facilitate controlled and protected release of model biologic (Bovine Serum Albumin), and anatomically-unhindered implantation under the acromion in a rodent model of acute rotator cuff repair. The matrix obtained from the highest polymeric-film casting concentration showed a controlled release of model biologics payload. The tested matrices rapidly degraded during the initial 4 weeks due to preferential hydrolysis of the lactide-rich regions within the polymer, and subsequently maintained a stable molecular weight due to the emergence of highly-crystalline caprolactone-rich regions. pH evaluation in the interior of the matrix showed minimal change signifying lesser accumulation of acidic degradation byproducts than seen in other bulk-degrading polymers, and maintenance of conformational stability of the model biologic payload. The context-dependent biocompatibility evaluation in a rodent model of acute rotator cuff repair showed matrix remodeling without eliciting excessive inflammatory reaction and is anticipated to completely degrade within 6 months. The engineered PLA-CL matrices offer unique advantages in controlled and protected biologic delivery, non-toxic biodegradation, and biocompatibility overcoming several limitations of commonly-used biodegradable polyesters.
Collapse
Affiliation(s)
- Anupama Prabhath
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut, USA
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, USA
- Department of Biomedical Engineering, UConn Health, Farmington, Connecticut, USA
| | - Varadraj N Vernekar
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut, USA
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, USA
| | - Vignesh Vasu
- Department of Material Science and Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Mary Badon
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut, USA
| | - Jean-Emmanuel Avochinou
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut, USA
| | - Alexandru D Asandei
- Department of Material Science and Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Sangamesh G Kumbar
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, USA
- Department of Biomedical Engineering, UConn Health, Farmington, Connecticut, USA
- Department of Material Science and Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Eckhard Weber
- Musculoskeletal Division, Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut, USA
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, USA
- Department of Biomedical Engineering, UConn Health, Farmington, Connecticut, USA
- Department of Material Science and Engineering, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
24
|
Geddes L, Themistou E, Burrows JF, Buchanan FJ, Carson L. Evaluation of the in vitro cytotoxicity and modulation of the inflammatory response by the bioresorbable polymers poly(D,L-lactide-co-glycolide) and poly(L-lactide-co-glycolide). Acta Biomater 2021; 134:261-275. [PMID: 34329786 DOI: 10.1016/j.actbio.2021.07.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/20/2022]
Abstract
Bioresorbable polymers composed of poly(D,L-lactide-co-glycolide) (PDLLGA) and poly(L-lactide-co-glycolide) (PLLGA) have become increasingly popular for the preparation of bone substitute constructs. However, there are reports of a delayed inflammatory reaction occurring months or years after implantation. Due to the long polymer degradation times, in vitro tests carried out at physiological temperature, 37°C, tend to assess only the short-term biocompatibility of these materials. The aim of this work is to develop an in vitro protocol that can be used to assess the long-term cytotoxicity of bioresorbable polymers in a time efficient manner. This study used a previously developed and validated accelerated degradation protocol to obtain samples of PDLLGA and PLLGA at increasing levels of degradation. Samples were then applied to standard ISO 10993-5 direct contact cytotoxicity testing and it was found that PDLLGA samples showed increasing levels of cytotoxicity at the later stages of degradation, with PLLGA samples demonstrating significantly less cytotoxic behaviour. Following concern that accumulation of acidic degradation products in a closed multi-well culture environment could overestimate cytotoxicity, we developed and validated a new dynamic flow culture methodology, for testing the cytotoxicity of these degradable materials, by adapting a commercial "organ on a chip" flow culture system, Quasi Vivo®. In addition to cytotoxicity testing, we have carried out profiling of inflammatory cytokines released by cells in response to degraded PDLLGA and PLLGA, and have suggested mechanism by which lactide-based bioresorbable materials could modulate the inflammatory response through the G-protein coupled receptor (GPCR), hydroxycarboxylic acid receptor 1 (HCA1). STATEMENT OF SIGNIFICANCE: Bioresorbable materials naturally disintegrate over time when implanted into the body. They are often used to make screws and clips for repair of broken bones. Unfortunately, some patients can react badly to the material, resulting in painful inflammation. Biomaterials scientists are interested in developing materials that are more compatible with the body. However, it is very difficult to predict the long-term compatibility of bioresorbable materials in the lab. In our study, we have developed a method that will allow us to study the effects of the materials as they continue to break down. This will help us understand why the materials may cause inflammation, and will support research into the development of new and improved materials for bone repair.
Collapse
Affiliation(s)
- Lucy Geddes
- School of Mechanical and Aerospace Engineering, Queens University Belfast, Ashby Building, Belfast, BT9 5AG, Northern Ireland, UK
| | - Efrosyni Themistou
- School of Chemistry and Chemical Engineering, Queens University Belfast, David Keir Building, Belfast, BT9 5AG, Northern Ireland, UK
| | - James F Burrows
- School of Pharmacy, Queens University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Fraser J Buchanan
- School of Mechanical and Aerospace Engineering, Queens University Belfast, Ashby Building, Belfast, BT9 5AG, Northern Ireland, UK
| | - Louise Carson
- School of Pharmacy, Queens University Belfast, Belfast, BT9 7BL, Northern Ireland, UK.
| |
Collapse
|
25
|
Figueiredo L, Makhni EC, Dierks M, Ferreira FC, Finkelstein S. Early cost estimating model for new bioabsorbable orthopedic implant candidates: A theoretical study. J Mech Behav Biomed Mater 2021; 124:104731. [PMID: 34500353 DOI: 10.1016/j.jmbbm.2021.104731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/14/2021] [Accepted: 07/17/2021] [Indexed: 11/24/2022]
Abstract
An early health technology assessment (HTA) study was performed to assess the need for developing a new bioabsorbable implant for the treatment of specific orthopedic injuries. The Anterior Cruciate Ligament Reconstruction (ACLR) procedure was selected based on the need and potential impact of bioabsorbable implants in the treatment of ACL injuries. The economic model considers the possible health events after an ACLR (failures and other complications such as stiffness and pain). A decision tree approach was used, and several sensitivity analyses were performed using a Monte Carlo simulation. A cost estimating model was applied comparatively for currently available metal and bioabsorbable implants against a potential new bioabsorbable implant with improved performance. A reduction in stiffness and pain symptoms were considered as targets for these new implants performance, with reduced inflammation resulting from the use of materials with appropriate biological and mechanical properties. The current study estimates that, under the assumptions made, the introduction of a new bioabsorbable implant in ACLR surgeries may generate yearly cost savings. The model estimates positive cost-benefits of the new implant when it reduces the probability of failure by more than 30%, or reduces at least 14% the probability of complications of an inflammatory nature. The development of a new bioabsorbable orthopedic implant for ACLR is encouraged by this study identifying the need for new bioabsorbable implants with improved biological and mechanical performance. The results of this early HTA have made it possible to anticipate design needs and objectives for the research and development of new orthopedic bioabsorbable implants.
Collapse
Affiliation(s)
- Lígia Figueiredo
- Institute of Mechanical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001, Lisboa, Portugal
| | - Eric C Makhni
- Department of Orthopedic Surgery, Henry Ford Health Systems, 2799 W Grand Blvd, Detroit, MI, 48202, United States
| | - Meghan Dierks
- Division of Clinical Informatics, Beth Israel Deaconess Medical Center, 1330 Beacon St., Suite 400, Brookline, MA, 02446, United States
| | - Frederico Castelo Ferreira
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001, Lisboa, Portugal; Associate Laboratory I4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
| | - Stan Finkelstein
- Institute for Data, Systems and Society, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, United States.
| |
Collapse
|
26
|
Mei X, Ye D, Zhang F, Di C. Implantable application of polymer‐based biosensors. JOURNAL OF POLYMER SCIENCE 2021. [DOI: 10.1002/pol.20210543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Xiangyuan Mei
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids Institute of Chemistry, Chinese Academy of Sciences Beijing China
- School of Chemical Sciences University of Chinese Academy of Sciences Beijing China
| | - Dekai Ye
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids Institute of Chemistry, Chinese Academy of Sciences Beijing China
| | - Fengjiao Zhang
- School of Chemical Sciences University of Chinese Academy of Sciences Beijing China
| | - Chong‐an Di
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids Institute of Chemistry, Chinese Academy of Sciences Beijing China
| |
Collapse
|
27
|
Shin EY, Kim DS, Lee MJ, Lee AR, Shim SH, Baek SW, Han DK, Lee DR. Prevention of chemotherapy-induced premature ovarian insufficiency in mice by scaffold-based local delivery of human embryonic stem cell-derived mesenchymal progenitor cells. Stem Cell Res Ther 2021; 12:431. [PMID: 34332643 PMCID: PMC8325282 DOI: 10.1186/s13287-021-02479-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/27/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is one of the most serious side effects of chemotherapy in young cancer survivors. It may not only reduce fecundity but also affect lifelong health. There is no standard therapy for preserving ovarian health after chemotherapy. Recently, administration of embryonic stem cell-derived mesenchymal progenitor cells (ESC-MPCs) has been considered a new therapeutic option for preventing POI. However, the previous method of directly injecting cells into the veins of patients exhibits low efficacy and safety. This study aimed to develop safe and effective local delivery methods for the prevention of POI using two types of bioinspired scaffolds. METHODS Female mice received intraperitoneal cisplatin for 10 days. On day 11, human ESC-MPCs were delivered through systemic administration using intravenous injection or local administration using intradermal injection and intradermal transplantation with a PLGA/MH sponge or hyaluronic acid (HA) gel (GEL) type of scaffold. PBS was injected intravenously as a negative control. Ovarian function and fertility were evaluated 4 weeks after transplantation. Follicle development was observed using hematoxylin and eosin staining. The plasma levels of sex hormones were measured using ELISA. Expression levels of anti-Müllerian hormone (AMH) and ki-67 were detected using immunostaining, and the quality of oocytes and embryos was evaluated after in vitro fertilization. The estrous cycles were observed at 2 months after transplantation. RESULTS The local administration of human ESC-MPCs using the bioinspired scaffold to the backs of mice effectively prolonged the cell survival rate in vivo. The HA GEL group exhibited the best recovered ovarian functions, including a significantly increased number of ovarian reserves, estrogen levels, and AMH levels and decreased apoptotic levels. Furthermore, the HA GEL group showed improved quality of oocytes and embryos and estrous cycle regularity. CONCLUSIONS HA GEL scaffolds can be used as new delivery platforms for ESC-MPC therapy, and this method may provide a novel option for the clinical treatment of chemotherapy-induced POI.
Collapse
Affiliation(s)
- Eun-Young Shin
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea
| | - Da-Seul Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Min Ji Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea
| | - Ah Reum Lee
- CHA Advanced Research Institute, CHA Medical Center, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea
| | - Sung Han Shim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea
| | - Seung Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea.
| | - Dong Ryul Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi, 13488, Republic of Korea.
| |
Collapse
|
28
|
Promotion of Bone Regeneration Using Bioinspired PLGA/MH/ECM Scaffold Combined with Bioactive PDRN. MATERIALS 2021; 14:ma14154149. [PMID: 34361342 PMCID: PMC8348682 DOI: 10.3390/ma14154149] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/26/2021] [Accepted: 07/22/2021] [Indexed: 12/16/2022]
Abstract
Current approaches of biomaterials for the repair of critical-sized bone defects still require immense effort to overcome numerous obstacles. The biodegradable polymer-based scaffolds have been required to expand further function for bone tissue engineering. Poly(lactic-co-glycolic) acid (PLGA) is one of the most common biopolymers owing to its biodegradability for tissue regenerations. However, there are major clinical challenges that the byproducts of the PLGA cause an acidic environment of implanting site. The critical processes in bone repair are osteogenesis, angiogenesis, and inhibition of excessive osteoclastogenesis. In this study, the porous PLGA (P) scaffold was combined with magnesium hydroxide (MH, M) and bone-extracellular matrix (bECM, E) to improve anti-inflammatory ability and osteoconductivity. Additionally, the bioactive polydeoxyribonucleotide (PDRN, P) was additionally incorporated in the existing PME scaffold. The prepared PMEP scaffold has pro-osteogenic and pro-angiogenic effects and inhibition of osteoclast due to the PDRN, which interacts with the adenosine A2A receptor agonist that up-regulates expression of vascular endothelial growth factor (VEGF) and down-regulates inflammatory cytokines. The PMEP scaffold has superior biological properties for human bone-marrow mesenchymal stem cells (hBMSCs) adhesion, proliferation, and osteogenic differentiation in vitro. Moreover, the gene expressions related to osteogenesis and angiogenesis of hBMSCs increased and the inflammatory factors decreased on the PMEP scaffold. In conclusion, it provides a promising strategy and clinical potential candidate for bone tissue regeneration and repairing bone defects.
Collapse
|
29
|
Ali Akbari Ghavimi S, Gehret PM, Aronson MR, Schipani R, Smith KWY, Borek RC, Germiller JA, Jacobs IN, Zur KB, Gottardi R. Drug delivery to the pediatric upper airway. Adv Drug Deliv Rev 2021; 174:168-189. [PMID: 33845038 DOI: 10.1016/j.addr.2021.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 11/25/2022]
Abstract
Pediatric upper airway disorders are frequently life-threatening and require precise assessment and intervention. Targeting these pathologies remains a challenge for clinicians due to the high complexity of pediatric upper airway anatomy and numerous potential etiologies; the most common treatments include systemic delivery of high dose steroids and antibiotics or complex and invasive surgeries. Furthermore, the majority of innovative airway management technologies are only designed and tested for adults, limiting their widespread implementation in the pediatric population. Here, we provide a comprehensive review of the most recent challenges of managing common pediatric upper airway disorders, describe the limitations of current clinical treatments, and elaborate on how to circumvent those limitations via local controlled drug delivery. Furthermore, we propose future advancements in the field of drug-eluting technologies to improve pediatric upper airway management outcomes.
Collapse
Affiliation(s)
- Soheila Ali Akbari Ghavimi
- Department of Surgery, Division of Otolaryngology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paul M Gehret
- Department of Surgery, Division of Otolaryngology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew R Aronson
- Department of Surgery, Division of Otolaryngology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rossana Schipani
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Kyra W Y Smith
- Department of Surgery, Division of Otolaryngology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ryan C Borek
- Department of Surgery, Division of Otolaryngology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - John A Germiller
- Department of Surgery, Division of Otolaryngology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Otorhinolaryngology - Head and Neck Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian N Jacobs
- Department of Surgery, Division of Otolaryngology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Otorhinolaryngology - Head and Neck Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karen B Zur
- Department of Surgery, Division of Otolaryngology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Otorhinolaryngology - Head and Neck Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Riccardo Gottardi
- Department of Surgery, Division of Otolaryngology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pediatrics, Division of Pulmonary Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Ri.MED Foundation, Palermo, PA 90133, Italy.
| |
Collapse
|
30
|
Behera M, Rajput M, Acharya S, Nadammal N, Suwas S, Chatterjee K. Zinc and cerium synergistically enhance the mechanical properties, corrosion resistance, and osteogenic activity of magnesium as resorbable biomaterials. Biomed Mater 2021; 16. [PMID: 34030150 DOI: 10.1088/1748-605x/ac0453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 05/24/2021] [Indexed: 11/11/2022]
Abstract
Magnesium and its alloys have the potential to serve as a revolutionary class of biodegradable materials, specifically in the field of degradable implants for orthopedics. However, the corrosion rate of commercially pure magnesium is high and does not match the rate of regeneration of bone tissues. In this work, magnesium alloys containing zinc and cerium, either alone or in combination, were investigated and compared with commercially-pure magnesium as biomaterials. The microstructure, mechanical properties, corrosion resistance, and response of osteoblastsin vitrowere systematically assessed. Results reveal that alloying with Ce results in grain refinement and weakening of texture. The tensile test revealed that the ternary alloy offered the best combination of elastic modulus (41.1 ± 0.5 GPa), tensile strength (234.5 ± 4.5 MPa), and elongation to break (17.1 ± 0.4%). The ternary alloy was also the most resistant to corrosion (current of 0.85 ± 0.05 × 10-4A cm-2) in simulated body fluid than the other alloys. The response of MC3T3-E1 cellsin vitrorevealed that the ternary alloy imparts minimal cytotoxicity. Interestingly, the ternary alloy was highly efficient in supporting osteogenic differentiation, as revealed by the expression of alkaline phosphatase and calcium deposition. In summary, the extruded Mg alloy containing both Zn and Ce exhibits a combination of mechanical properties, corrosion resistance, and cell response that is highly attractive for engineering biodegradable orthopedic implants.
Collapse
Affiliation(s)
- Manisha Behera
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore, 560012, India
| | - Monika Rajput
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore, 560012, India
| | - Srijan Acharya
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore, 560012, India
| | - Naresh Nadammal
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore, 560012, India
| | - Satyam Suwas
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore, 560012, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore, 560012, India
| |
Collapse
|
31
|
Research status of biodegradable metals designed for oral and maxillofacial applications: A review. Bioact Mater 2021; 6:4186-4208. [PMID: 33997502 PMCID: PMC8099919 DOI: 10.1016/j.bioactmat.2021.01.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/10/2021] [Accepted: 01/10/2021] [Indexed: 01/08/2023] Open
Abstract
The oral and maxillofacial regions have complex anatomical structures and different tissue types, which have vital health and aesthetic functions. Biodegradable metals (BMs) is a promising bioactive materials to treat oral and maxillofacial diseases. This review summarizes the research status and future research directions of BMs for oral and maxillofacial applications. Mg-based BMs and Zn-based BMs for bone fracture fixation systems, and guided bone regeneration (GBR) membranes, are discussed in detail. Zn-based BMs with a moderate degradation rate and superior mechanical properties for GBR membranes show great potential for clinical translation. Fe-based BMs have a relatively low degradation rate and insoluble degradation products, which greatly limit their application and clinical translation. Furthermore, we proposed potential future research directions for BMs in the oral and maxillofacial regions, including 3D printed BM bone scaffolds, surface modification for BMs GBR membranes, and BMs containing hydrogels for cartilage regeneration, soft tissue regeneration, and nerve regeneration. Taken together, the progress made in the development of BMs in oral and maxillofacial regions has laid a foundation for further clinical translation.
Collapse
|
32
|
Chen YC, Gad SF, Chobisa D, Li Y, Yeo Y. Local drug delivery systems for inflammatory diseases: Status quo, challenges, and opportunities. J Control Release 2021; 330:438-460. [PMID: 33352244 DOI: 10.1016/j.jconrel.2020.12.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
Inflammation that is not resolved in due course becomes a chronic disease. The treatment of chronic inflammatory diseases involves a long-term use of anti-inflammatory drugs such as corticosteroids and nonsteroidal anti-inflammatory drugs, often accompanied by dose-dependent side effects. Local drug delivery systems have been widely explored to reduce their off-target side effects and the medication frequency, with several products making to the market or in development over the years. However, numerous challenges remain, and drug delivery technology is underutilized in some applications. This review showcases local drug delivery systems in different inflammatory diseases, including the targets well-known to drug delivery scientists (e.g., joints, eyes, and teeth) and other applications with untapped opportunities (e.g., sinus, bladder, and colon). In each section, we start with a brief description of the disease and commonly used therapy, introduce local drug delivery systems currently on the market or in the development stage, focusing on polymeric systems, and discuss the remaining challenges and opportunities in future product development.
Collapse
Affiliation(s)
- Yun-Chu Chen
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Sheryhan F Gad
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Dhawal Chobisa
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Integrated product development organization, Innovation plaza, Dr. Reddy's Laboratories, Hyderabad 500090, India
| | - Yongzhe Li
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
33
|
Magnetron sputtered magnesium-based thin film metallic glasses for bioimplants. Biointerphases 2021; 16:011005. [PMID: 33706531 DOI: 10.1116/6.0000535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Mg-based thin film metallic glasses (TFMGs) can viably decrease stress shielding caused by mismatch of the modulus of elasticity between the implant material and human bone. Here, Mg-based TFMGs were fabricated onto implantable substrates by ion assisted pulsed DC magnetron sputtering. The microstructure assessment and the impact of the principle constituents of the coatings were determined utilizing an x-ray diffractometer, a transmission electron microscope, and x-ray photoelectron spectroscopy. The hardness of these thin films was estimated to be 5.1 GPa. In vitro degradation tests including electrochemical studies and immersion tests in simulated body fluid revealed that the presence of zinc could raise the corrosion resistance of Mg-based TFMG. Indirect in vitro cytotoxicity using L929 fibroblast cells revealed that the TFMGs did not induce any toxicity in cells. Biomineralization experiments using Saos-2 cells promoted the formation of calcium phosphate on its surface.
Collapse
|
34
|
Characteristics of the Mg-Zn-Ca-Gd Alloy after Mechanical Alloying. MATERIALS 2021; 14:ma14010226. [PMID: 33466438 PMCID: PMC7796461 DOI: 10.3390/ma14010226] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/28/2020] [Accepted: 12/31/2020] [Indexed: 12/31/2022]
Abstract
Magnesium-based materials are interesting alternatives for medical implants, as they have promising mechanical and biological properties. Thanks to them, it is possible to create biodegradable materials for medical application, which would reduce both costs and time of treatment. Magnesium as the sole material, however, it is not enough to support this function. It is important to determine proper alloying elements and methods. A viable method for creating such alloys is mechanical alloying, which can be used to design the structure and properties for proper roles. Mechanical alloying is highly influenced by the milling time of the alloy, as the time of the process affects many properties of the milled powders. X-ray diffraction (XRD) and scanning electron microscopy (SEM) with energy-dispersive spectroscopy (EDS) were carried out to study the powder morphology and chemical composition of Mg65Zn30Ca4Gd1 powders. Moreover, the powder size was assessed by granulometric method and the Vickers hardness test was used for microhardness testing. The samples were milled for 6 min, 13, 20, 30, 40, and 70 h. The hardness correlated with the particle size of the samples. After 30 h of milling time, the average value of hardness was equal to 168 HV and it was lower after 13 (333 HV), 20 (273 HV), 40 (329 HV), and 70 (314 HV) h. The powder particles average size increased after 13 (31 μm) h of milling time, up to 30 (45–49 μm) hours, and then sharply decreased after 40 (28 μm) and 70 (12 μm) h.
Collapse
|
35
|
Singh R, Bathaei MJ, Istif E, Beker L. A Review of Bioresorbable Implantable Medical Devices: Materials, Fabrication, and Implementation. Adv Healthc Mater 2020; 9:e2000790. [PMID: 32790033 DOI: 10.1002/adhm.202000790] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/22/2020] [Indexed: 12/15/2022]
Abstract
Implantable medical devices (IMDs) are designed to sense specific parameters or stimulate organs and have been actively used for treatment and diagnosis of various diseases. IMDs are used for long-term disease screening or treatments and cannot be considered for short-term applications since patients need to go through a surgery for retrieval of the IMD. Advances in bioresorbable materials has led to the development of transient IMDs that can be resorbed by bodily fluids and disappear after a certain period. These devices are designed to be implanted in the adjacent of the targeted tissue for predetermined times with the aim of measurement of pressure, strain, or temperature, while the bioelectronic devices stimulate certain tissues. They enable opportunities for monitoring and treatment of acute diseases. To realize such transient and miniaturized devices, researchers utilize a variety of materials, novel fabrication methods, and device design strategies. This review discusses potential bioresorbable materials for each component in an IMD followed by programmable degradation and safety standards. Then, common fabrication methods for bioresorbable materials are introduced, along with challenges. The final section provides representative examples of bioresorbable IMDs for various applications with an emphasis on materials, device functionality, and fabrication methods.
Collapse
Affiliation(s)
- Rahul Singh
- Department of Mechanical Engineering Koç University Rumelifeneri Yolu, Sarıyer Istanbul 34450 Turkey
| | - Mohammad Javad Bathaei
- Department of Biomedical Sciences and Engineering Koç University Rumelifeneri Yolu, Sarıyer Istanbul 34450 Turkey
| | - Emin Istif
- Department of Mechanical Engineering Koç University Rumelifeneri Yolu, Sarıyer Istanbul 34450 Turkey
| | - Levent Beker
- Department of Mechanical Engineering Koç University Rumelifeneri Yolu, Sarıyer Istanbul 34450 Turkey
| |
Collapse
|
36
|
Lu X, Miao L, Gao W, Chen Z, McHugh KJ, Sun Y, Tochka Z, Tomasic S, Sadtler K, Hyacinthe A, Huang Y, Graf T, Hu Q, Sarmadi M, Langer R, Anderson DG, Jaklenec A. Engineered PLGA microparticles for long-term, pulsatile release of STING agonist for cancer immunotherapy. Sci Transl Med 2020; 12:eaaz6606. [PMID: 32801144 PMCID: PMC9019818 DOI: 10.1126/scitranslmed.aaz6606] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/06/2020] [Accepted: 06/29/2020] [Indexed: 08/02/2023]
Abstract
Activation of the stimulator of interferon gene (STING) pathway within the tumor microenvironment has been shown to generate a strong antitumor response. Although local administration of STING agonists has promise for cancer immunotherapy, the dosing regimen needed to achieve efficacy requires frequent intratumoral injections over months. Frequent dosing for cancer treatment is associated with poor patient adherence, with as high as 48% of patients failing to comply. Multiple intratumoral injections also disrupt the tumor microenvironment and vascular networks and therefore increase the risk of metastasis. Here, we developed microfabricated polylactic-co-glycolic acid (PLGA) particles that remain at the site of injection and release encapsulated STING agonist as a programmable sequence of pulses at predetermined time points that mimic multiple injections over days to weeks. A single intratumoral injection of STING agonist-loaded microparticles triggered potent local and systemic antitumor immune responses, inhibited tumor growth, and prolonged survival as effectively as multiple soluble doses, but with reduced metastasis in several mouse tumor models. STING agonist-loaded microparticles improved the response to immune checkpoint blockade therapy and substantially decreased the tumor recurrence rate from 100 to 25% in mouse models of melanoma when administered during surgical resection. In addition, we demonstrated the therapeutic efficacy of STING microparticles on an orthotopic pancreatic cancer model in mice that does not allow multiple intratumoral injections. These findings could directly benefit current STING agonist therapy by decreasing the number of injections, reducing risk of metastasis, and expanding its applicability to hard-to-reach cancers.
Collapse
Affiliation(s)
- Xueguang Lu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lei Miao
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wenting Gao
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ziqi Chen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kevin J McHugh
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, TX 77005, USA
| | - Yehui Sun
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Zachary Tochka
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stephanie Tomasic
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kaitlyn Sadtler
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Section on Immuno-Engineering, National Institute for Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20894, USA
| | - Alain Hyacinthe
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yuxuan Huang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tyler Graf
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Quanyin Hu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Morteza Sarmadi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ana Jaklenec
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
37
|
Patel A, Zaky SH, Schoedel K, Li H, Sant V, Beniash E, Sfeir C, Stolz DB, Sant S. Design and evaluation of collagen-inspired mineral-hydrogel nanocomposites for bone regeneration. Acta Biomater 2020; 112:262-273. [PMID: 32497742 DOI: 10.1016/j.actbio.2020.05.034] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/29/2020] [Accepted: 05/26/2020] [Indexed: 12/21/2022]
Abstract
Bone loss due to trauma and tumors remains a serious clinical concern. Due to limited availability and disease transmission risk with autografts and allografts, calcium phosphate bone fillers and growth factor-based substitute bone grafts are currently used in the clinic. However, substitute grafts lack bone regeneration potential when used without growth factors. When used along with the added growth factors, they lead to unwanted side effects such as uncontrolled bone growth. Collagen-based hydrogel grafts available on the market fail to provide structural guidance to native cells due to high water-solubility and faster degradation. To overcome these limitations, we employed bioinspired material design and fabricated three different hydrogels with structural features similar to native collagen at multiple length-scales. These hydrogels fabricated using polyionic complexation of oppositely charged natural polysaccharides exhibited multi-scale architecture mimicking nanoscale banding pattern, and microscale fibrous structure of native collagen. All three hydrogels promoted biomimetic apatite-like mineral deposition in vitro elucidating crystalline structure on the surface while amorphous calcium phosphate inside the hydrogels resulting in mineral-hydrogel nanocomposites. When evaluated in a non-load bearing critical size mouse calvaria defect model, chitosan - kappa carrageenan mineral-hydrogel nanocomposites enhanced bone regeneration without added growth factors compared to empty defect as well as widely used marketed collagen scaffolds. Histological assessment of the regenerated bone revealed improved healing and tissue remodeling with mineral-hydrogel nanocomposites. Overall, these collagen-inspired mineral-hydrogel nanocomposites showed multi-scale hierarchical structure and can potentially serve as promising bioactive hydrogel to promote bone regeneration. STATEMENT OF SIGNIFICANCE: Hydrogels, especially collagen, are widely used in bone tissue engineering. Collagen fibrils play arguably the most important role during natural bone development. Its multi-scale hierarchical structure to form fibers from fibrils and electrostatic charges enable mineral sequestration, nucleation, and growth. However, bulk collagen hydrogels exhibit limited bone regeneration and are mostly used as carriers for highly potent growth factors such as bone morphogenic protein-2, which increase the risk of uncontrolled bone growth. Thus, there is an unmet clinical need for a collagen-inspired biomaterial that can recreate structural hierarchy, mineral sequestration ability, and stimulate recruitment of host progenitor cells to facilitate bone regeneration. Here, we propose collagen-inspired bioactive mineral-hydrogel nanocomposites as a growth factor-free approach to guide and enhance bone regeneration.
Collapse
Affiliation(s)
- Akhil Patel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261
| | - Samer H Zaky
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Karen Schoedel
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Hongshuai Li
- Musculoskeletal Growth & Regeneration Laboratory, Department of Orthopaedic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Vinayak Sant
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261
| | - Elia Beniash
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15261; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15219; McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15260
| | - Charles Sfeir
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15261; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15219; McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15260
| | - Donna B Stolz
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261; McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15260; Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15260
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15219; McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15260; UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15260.
| |
Collapse
|
38
|
Wang C, Ye X, Zhao Y, Bai L, He Z, Tong Q, Xie X, Zhu H, Cai D, Zhou Y, Lu B, Wei Y, Mei L, Xie D, Wang M. Cryogenic 3D printing of porous scaffolds for in situ delivery of 2D black phosphorus nanosheets, doxorubicin hydrochloride and osteogenic peptide for treating tumor resection-induced bone defects. Biofabrication 2020; 12:035004. [PMID: 31952065 DOI: 10.1088/1758-5090/ab6d35] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor resection is widely used to prevent tumor growth. However, the defected tissue at the original tumor site also causes tissue or organ dysfunction which lowers the patient's life quality. Therefore, regenerating the tissue and preventing tumor recurrence are highly important. Herein, according to the concept of 'first kill and then regenerate', a versatile scaffold-based tissue engineering strategy based on cryogenic 3D printing of water-in-oil polyester emulsion inks, containing multiple functional agents, was developed, in order to realize the elimination of tumor cells with recurrence suppression and improved tissue regeneration sequentially. To illustrate our strategy, water/poly(lactic-co-glycolic acid)/dichloromethane emulsions containing β-tricalcium phosphate (β-TCP), 2D black phosphorus (BP) nanosheets, low-dose doxorubicin hydrochloride (DOX) and high-dose osteogenic peptide were cryogenically 3D printed into hierarchically porous and mechanically strong nanocomposite scaffolds, with multiple functions to treat bone tumor, resection-induced tissue defects. Prompt tumor ablation and long-term suppression of tumor recurrence could be achieved due to the synergistic effects of photothermotherapy and chemotherapy, and improved bone regeneration was obtained eventually due to the presence of bony environment and sustained peptide release. Notably, BP nanosheets in scaffolds significantly reduced the long-term toxicity phenomenon of released DOX during in vivo bone regeneration. Our study also provides insights for the design of multi-functional tissue engineering scaffolds for treating other tumor resection-induced tissue defects.
Collapse
Affiliation(s)
- Chong Wang
- College of Mechanical Engineering, Dongguan University of Technology, Dongguan, Guangdong, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hameister R, Kaur C, Dheen ST, Lohmann CH, Singh G. Reactive oxygen/nitrogen species (ROS/RNS) and oxidative stress in arthroplasty. J Biomed Mater Res B Appl Biomater 2020; 108:2073-2087. [PMID: 31898397 DOI: 10.1002/jbm.b.34546] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/19/2019] [Accepted: 12/08/2019] [Indexed: 12/16/2022]
Abstract
The interplay between implant design, biomaterial characteristics, and the local microenvironment adjacent to the implant is of utmost importance for implant performance and success of the joint replacement surgery. Reactive oxygen and nitrogen species (ROS/RNS) are among the various factors affecting the host as well as the implant components. Excessive formation of ROS and RNS can lead to oxidative stress, a condition that is known to damage cells and tissues and also to affect signaling pathways. It may further compromise implant longevity by accelerating implant degradation, primarily through activation of inflammatory cells. In addition, wear products of metallic, ceramic, polyethylene, or bone cement origin may also generate oxidative stress themselves. This review outlines the generation of free radicals and oxidative stress in arthroplasty and provides a conceptual framework on its implications for soft tissue remodeling and bone resorption (osteolysis) as well as implant longevity. Key findings derived from cell culture studies, animal models, and patients' samples are presented. Strategies to control oxidative stress by implant design and antioxidants are explored and areas of controversy and challenges are highlighted. Finally, directions for future research are identified. A better understanding of the host-implant interplay and the role of free radicals and oxidative stress will help to evaluate therapeutic approaches and will ultimately improve implant performance in arthroplasty.
Collapse
Affiliation(s)
- Rita Hameister
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shaikali Thameem Dheen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christoph H Lohmann
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Gurpal Singh
- Centre for Orthopaedics Pte Ltd, Singapore, Singapore
| |
Collapse
|
40
|
Wang G, Jiang W, Mo S, Xie L, Liao Q, Hu L, Ruan Q, Tang K, Mehrjou B, Liu M, Tong L, Wang H, Zhuang J, Wu G, Chu PK. Nonleaching Antibacterial Concept Demonstrated by In Situ Construction of 2D Nanoflakes on Magnesium. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902089. [PMID: 31921567 PMCID: PMC6947590 DOI: 10.1002/advs.201902089] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Indexed: 05/19/2023]
Abstract
In bone implants, antibacterial biomaterials with nonleaching surfaces are superior to ones based on abrupt release because systemic side effects arising from the latter can be avoided. In this work, a nonleaching antibacterial concept is demonstrated by fabricating 2D nanoflakes in situ on magnesium (Mg). Different from the conventional antibacterial mechanisms that depend on Mg2+ release and pH increase, the nanoflakes exert mechanical tension onto the bacteria membranes to destroy microorganisms on contact and produce intracellular stress via physical interactions, which is also revealed by computational simulations. Moreover, the nanoflake layer decelerates the corrosion process resulting in mitigated Mg2+ release, weaker alkalinity in the vicinity, and less hydrogen evolution, in turn inducing less inflammatory reactions and ensuring the biocompatibility as confirmed by the in vivo study. In this way, bacteria are killed by a mechanical process causing very little side effects. This work provides information and insights pertaining to the design of multifunctional biomaterials.
Collapse
Affiliation(s)
- Guomin Wang
- Department of PhysicsDepartment of Materials Science and Engineeringand Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong KongChina
| | - Wenjuan Jiang
- College of PharmacyWestern University of Health Sciences309 E. Second StPomonaCA91766USA
| | - Shi Mo
- Department of PhysicsDepartment of Materials Science and Engineeringand Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong KongChina
| | - Lingxia Xie
- Research Center for Biomedical Materials and InterfacesShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Qing Liao
- Research Center for Biomedical Materials and InterfacesShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Liangsheng Hu
- Department of Chemistry and Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong ProvinceShantou UniversityGuangdong515063P. R. China
| | - Qingdong Ruan
- Department of PhysicsDepartment of Materials Science and Engineeringand Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong KongChina
| | - Kaiwei Tang
- Department of PhysicsDepartment of Materials Science and Engineeringand Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong KongChina
| | - Babak Mehrjou
- Department of PhysicsDepartment of Materials Science and Engineeringand Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong KongChina
| | - Mengting Liu
- USC Stevens Neuroimaging and Informatics InstituteKeck School of Medicine of USCUniversity of Southern CaliforniaLos AngelesCA90033USA
| | - Liping Tong
- Research Center for Biomedical Materials and InterfacesShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Huaiyu Wang
- Research Center for Biomedical Materials and InterfacesShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Jie Zhuang
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of SciencesSuzhou215163P. R. China
| | - Guosong Wu
- College of Mechanics and MaterialsHohai UniversityNanjing211100P. R. China
| | - Paul K. Chu
- Department of PhysicsDepartment of Materials Science and Engineeringand Department of Biomedical EngineeringCity University of Hong KongTat Chee AvenueKowloonHong KongChina
| |
Collapse
|
41
|
Deshmukh K, Kovářík T, Křenek T, Docheva D, Stich T, Pola J. Recent advances and future perspectives of sol–gel derived porous bioactive glasses: a review. RSC Adv 2020; 10:33782-33835. [PMID: 35519068 PMCID: PMC9056785 DOI: 10.1039/d0ra04287k] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/02/2020] [Indexed: 12/22/2022] Open
Abstract
Sol–gel derived bioactive glasses have been extensively explored as a promising and highly porous scaffold materials for bone tissue regeneration applications owing to their exceptional osteoconductivity, osteostimulation and degradation rates.
Collapse
Affiliation(s)
- Kalim Deshmukh
- New Technologies – Research Center
- University of West Bohemia
- Plzeň
- Czech Republic
| | - Tomáš Kovářík
- New Technologies – Research Center
- University of West Bohemia
- Plzeň
- Czech Republic
| | - Tomáš Křenek
- New Technologies – Research Center
- University of West Bohemia
- Plzeň
- Czech Republic
| | - Denitsa Docheva
- Experimental Trauma Surgery
- Department of Trauma Surgery
- University Regensburg Medical Centre
- Regensburg
- Germany
| | - Theresia Stich
- Experimental Trauma Surgery
- Department of Trauma Surgery
- University Regensburg Medical Centre
- Regensburg
- Germany
| | - Josef Pola
- New Technologies – Research Center
- University of West Bohemia
- Plzeň
- Czech Republic
| |
Collapse
|
42
|
Obeid BA. Implants and grafts used in fractures for early healing. JOURNAL OF ORTHOPAEDICS AND SPINE 2020. [DOI: 10.4103/joas.joas_45_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
43
|
Cortez Alcaraz MC, Cipriano AF, Lin J, Soria P, Tian Q, Liu H. Electrophoretic Deposition of Magnesium Oxide Nanoparticles on Magnesium: Processing Parameters, Microstructures, Degradation, and Cytocompatibility. ACS APPLIED BIO MATERIALS 2019; 2:5634-5652. [PMID: 35021558 DOI: 10.1021/acsabm.9b00714] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Magnesium (Mg) and its alloys are a class of promising materials for biodegradable orthopedic and craniomaxillofacial implants; however, rapid release of hydrogen gas remains a key challenge for clinical translation. This study reported the optimal parameters of electrophoretic deposition (EPD), at which magnesium oxide nanoparticles (nMgO) could be deposited onto Mg substrates with homogeneous surface morphology and elemental distribution. The results showed that the distribution and uniformity of the nMgO coatings on Mg improved when the nMgO concentration in ethanol increased and the time of applied voltage decreased. The nMgO-coated Mg showed a homogeneous surface and distinct degradation mode during the 9-day immersion studies in revised simulated body fluid (r-SBF) and Dulbecco's modified Eagle's medium (DMEM), when compared with the noncoated Mg controls. The nMgO coating initially mitigated hydrogen gas formation. The degradation layer on nMgO-coated Mg was thicker than the noncoated Mg and enriched with Ca and P that are favorable for skeletal implant applications. In the direct culture study with bone marrow derived mesenchymal stem cells (BMSCs) in vitro, the cell adhesion density and morphology were not affected by the solubilized degradation products released by the nMgO-coated Mg under indirect contact. However, at the cell-biomaterial interface, the cell spreading decreased under direct contact, possibly because of the continuous dynamic degradation of the samples. The electrophoretically deposited nMgO coatings on Mg-based medical implants should be further studied to improve the coating-substrate and cell-material interfaces for clinical applications.
Collapse
Affiliation(s)
| | | | | | - Pedro Soria
- Department of Biology, California State University, San Bernardino, California 92407, United States
| | | | | |
Collapse
|
44
|
Martins JA, Lach AA, Morris HL, Carr AJ, Mouthuy PA. Polydioxanone implants: A systematic review on safety and performance in patients. J Biomater Appl 2019; 34:902-916. [PMID: 31771403 PMCID: PMC7044756 DOI: 10.1177/0885328219888841] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Joana A Martins
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Antonina A Lach
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Hayley L Morris
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Andrew J Carr
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Pierre-Alexis Mouthuy
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
45
|
Cai H, Meng J, Li X, Xue F, Chu C, Guo C, Bai J. In vitro degradation behavior of Mg wire/poly(lactic acid) composite rods prepared by hot pressing and hot drawing. Acta Biomater 2019; 98:125-141. [PMID: 31146034 DOI: 10.1016/j.actbio.2019.05.059] [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: 11/25/2018] [Revised: 05/10/2019] [Accepted: 05/23/2019] [Indexed: 12/31/2022]
Abstract
In this study, we prepared Mg wire/poly(lactic acid) composite rods by hot pressing (HP) and hot drawing (HD) processes, which show desirable application potential as a biodegradable implant in orthopedics. The influences of the volume content of Mg wires and processing steps on the degradation behavior of composite rods as well as the mutual influence between Mg and poly(lactic acid) (PLA) during degradation were investigated. During degradation, the improved interface bonding between Mg and PLA by micro-arc oxidation can effectively inhibit the diffusion of the medium from the ends to the central part, supported by XR-CT results, which show the degradation cracks in PLA initiated and propagated along the radial direction of the rod rather than the axial direction. When compared with the hot pressed rod, the three passes hot drawn rod had lower degradation rate and better strength retention during long-time immersion. This is ascribed to the higher crystallinity of the PLA matrix by HD. For both hot pressed and hot drawn rods, more Mg content of the wires will result in an increase in the degradation rate of PLA due to alkaline catalytic hydrolysis. Therefore, we can not only predict the degradation period by the present degradation mathematical model but also further control degradation rate by adjusting processing steps and wire volume content. This will be helpful for future material design and indication selection. STATEMENT OF SIGNIFICANCE: We investigated the influence of interface microstructure, self-reinforced PLA matrix, and wire volume content on the degradation behavior and clarified the differences in the degradation mechanism of composite rods. The 3D morphology of cracks in composite rods enlightened us a clear understanding about the emergence and evolution of cracks, which revealed the reason for the failure of composite rods. The degradation model can be used to predict the degradation period and provide valuable information for their future application.
Collapse
|
46
|
Datye A, Alexander Kube S, Verma D, Schroers J, Schwarz UD. Accelerated discovery and mechanical property characterization of bioresorbable amorphous alloys in the Mg-Zn-Ca and the Fe-Mg-Zn systems using high-throughput methods. J Mater Chem B 2019; 7:5392-5400. [PMID: 31411619 DOI: 10.1039/c9tb01302d] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Ternary amorphous alloys in the magnesium (Mg)-zinc (Zn)-calcium (Ca) and the iron (Fe)-Mg-Zn systems are promising candidates for use in bioresorbable implants and devices. The optimal alloy compositions for biomedical applications should be chosen from a large variety of available alloys with best combination of mechanical properties (modulus, strength, hardness) and biological response (in situ degradation rates, cell adhesion and proliferation). As a first step towards establishing a database designed to enable such targeted material selection, amorphous alloy composition libraries were fabricated employing a combinatorial magnetron sputtering approach where Mg, Zn, and Ca/Fe are co-deposited from separate sources onto a silicon wafer substrate. Composition analysis using energy dispersive X-ray spectroscopy documented a composition range of ∼15-85 at% Mg, ∼6-55 at% Zn, and ∼5-60 at% Ca for the Mg-Zn-Ca library and ∼26-84 at% Mg, ∼10-61 at% Zn, and ∼7-55 at% Fe for the Fe-Mg-Zn library. X-ray diffraction measurements established that amorphous alloys (i.e., glasses) form in almost the entire range of composition at the high cooling rates during sputtering for both alloy libraries. Finally, the effective material modulus, the Oliver-Pharr hardness, and the yield strength values obtained using nanoindentation reveal a wide range of mechanical properties within both systems.
Collapse
Affiliation(s)
- Amit Datye
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06511, USA.
| | - Sebastian Alexander Kube
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06511, USA.
| | - Devendra Verma
- Nanoscience Instruments, 10008 S. 51st Street, Ste 110, Phoenix, AZ 85044, USA
| | - Jan Schroers
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06511, USA.
| | - Udo D Schwarz
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06511, USA. and Department of Chemical Engineering, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
47
|
Chen D, Weng L, Chen C, Zheng J, Wu T, Zeng S, Zhang S, Xiao J. Inflammation and dysfunction in human aortic endothelial cells associated with poly-l-lactic acid degradation in vitro are alleviated by curcumin. J Biomed Mater Res A 2019; 107:2756-2763. [PMID: 31408261 DOI: 10.1002/jbm.a.36778] [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: 10/30/2018] [Revised: 03/14/2019] [Accepted: 08/07/2019] [Indexed: 01/10/2023]
Abstract
Poly-l-lactic acid (PLLA) is widely used in clinic, for example, as biodegradable coronary artery stents. However, inflammatory responses in endothelial cells associated with PLLA degradation are relatively undefined. We previously reported inflammation in human aortic endothelial cells (HAEC) in vitro and in vivo. Here, we further assessed inflammatory injury, including cell migration, cell function, and inflammatory cytokines expressed in HAEC treated with PLLA and curcumin by CCK-8, wound healing assay, ELISA, and Western blot. Significant inhibition of cell migration, remarkable dysfunction, and inflammatory responses were found in HAEC treated with PLLA degradation extract, and these effects were alleviated by Cur treatment. These findings indicated that cautious evaluation of biodegradable polymers should be performed, and Cur represents a promising anti-inflammatory agent for alleviating endothelial dysfunction and inflammation caused by PLLA degradation. In addition, Cur should be further studied experimentally in in vivo experiments on animal models as a potential therapeutic to reduce thrombosis of biodegradable polymer stents.
Collapse
Affiliation(s)
- Dongping Chen
- Central Laboratory, The Dongguan Affiliated Hospital of Medical College of Jinan University, The Fifth People's Hospital of Dongguan, Dongguan, China
| | - Linsheng Weng
- Department of Cardiology, The Dongguan Affiliated Hospital of Medical College of Jinan University, The Fifth People's Hospital of Dongguan, Dongguan, China
| | - Can Chen
- Department of Pathology, The Dongguan Affiliated Hospital of Medical College of Jinan University, The Fifth People's Hospital of Dongguan, Dongguan, China
| | - Jian Zheng
- Dongguan TT Medical, Inc., Dongguan, China
| | - Tim Wu
- Dongguan TT Medical, Inc., Dongguan, China.,Vaso Tech, Inc., Lowell, Massachusetts
| | - Sufen Zeng
- Central Laboratory, The Dongguan Affiliated Hospital of Medical College of Jinan University, The Fifth People's Hospital of Dongguan, Dongguan, China
| | - Suzhen Zhang
- Central Laboratory, The Dongguan Affiliated Hospital of Medical College of Jinan University, The Fifth People's Hospital of Dongguan, Dongguan, China
| | - Jianmin Xiao
- Central Laboratory, The Dongguan Affiliated Hospital of Medical College of Jinan University, The Fifth People's Hospital of Dongguan, Dongguan, China.,Department of Cardiology, The Dongguan Affiliated Hospital of Medical College of Jinan University, The Fifth People's Hospital of Dongguan, Dongguan, China
| |
Collapse
|
48
|
Cha GD, Kang D, Lee J, Kim D. Bioresorbable Electronic Implants: History, Materials, Fabrication, Devices, and Clinical Applications. Adv Healthc Mater 2019; 8:e1801660. [PMID: 30957984 DOI: 10.1002/adhm.201801660] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 02/14/2019] [Indexed: 12/13/2022]
Abstract
Medical implants, either passive implants for structural support or implantable devices with active electronics, have been widely used for the diagnosis and treatment of various diseases and clinical issues. These implants offer various functions, including mechanical support of biological structures in orthopedic and dental applications, continuous electrophysiological monitoring and feedback of electrical stimulation in neuronal and cardiac applications, and controlled drug delivery while maintaining arterial structure in drug-eluting stents. Although these implants exhibit long-term biocompatibility, surgery for their retrieval is often required, which imposes physical, biological, and economical burdens on the patients. Therefore, as an alternative to such secondary surgeries, bioresorbable implants that disappear after a certain period of time inside the body, including bioresorbable active electronics, have been highlighted recently. This review first discusses the historical background of medical implants and briefly define related terminology. Representative examples of non-degradable medical implants for passive structural support and/or for diagnosis and therapy with active electronics are also provided. Then, recent progress in bioresorbable active implants composed of biosignal sensors, actuators for therapeutics, wireless power supply components, and their integrated systems are reviewed. Finally, clinical applications of these bioresorbable electronic implants are exemplified with brief conclusion and future outlook.
Collapse
Affiliation(s)
- Gi Doo Cha
- Center for Nanoparticle ResearchInstitute for Basic Science (IBS) Seoul 08826 Republic of Korea
- School of Chemical and Biological EngineeringInstitute of Chemical ProcessesSeoul National University (SNU) Seoul 08826 Republic of Korea
| | - Dayoung Kang
- Center for Nanoparticle ResearchInstitute for Basic Science (IBS) Seoul 08826 Republic of Korea
- School of Chemical and Biological EngineeringInstitute of Chemical ProcessesSeoul National University (SNU) Seoul 08826 Republic of Korea
| | - Jongha Lee
- Center for Nanoparticle ResearchInstitute for Basic Science (IBS) Seoul 08826 Republic of Korea
- School of Chemical and Biological EngineeringInstitute of Chemical ProcessesSeoul National University (SNU) Seoul 08826 Republic of Korea
| | - Dae‐Hyeong Kim
- Center for Nanoparticle ResearchInstitute for Basic Science (IBS) Seoul 08826 Republic of Korea
- School of Chemical and Biological EngineeringInstitute of Chemical ProcessesSeoul National University (SNU) Seoul 08826 Republic of Korea
| |
Collapse
|
49
|
Lih E, Park W, Park KW, Chun SY, Kim H, Joung YK, Kwon TG, Hubbell JA, Han DK. A Bioinspired Scaffold with Anti-Inflammatory Magnesium Hydroxide and Decellularized Extracellular Matrix for Renal Tissue Regeneration. ACS CENTRAL SCIENCE 2019; 5:458-467. [PMID: 30937373 PMCID: PMC6439446 DOI: 10.1021/acscentsci.8b00812] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Indexed: 05/23/2023]
Abstract
Kidney diseases are a worldwide public health issue. Renal tissue regeneration using functional scaffolds with biomaterials has attracted a great deal of attention due to limited donor organ availability. Here, we developed a bioinspired scaffold that can efficiently induce renal tissue regeneration. The bioinspired scaffold was designed with poly(lactide-co-glycolide) (PLGA), magnesium hydroxide (Mg(OH)2), and decellularized renal extracellular matrix (ECM). The Mg(OH)2 inhibited materials-induced inflammatory reactions by neutralizing the acidic microenvironment formed by degradation products of PLGA, and the acellular ECM helped restore the biological function of kidney tissues. When the PLGA/ECM/Mg(OH)2 scaffold was implanted in a partially nephrectomized mouse model, it led to the regeneration of renal glomerular tissue with a low inflammatory response. Finally, the PLGA/ECM/Mg(OH)2 scaffold was able to restore renal function more effectively than the control groups. These results suggest that the bioinspired scaffold can be used as an advanced scaffold platform for renal disease treatment.
Collapse
Affiliation(s)
- Eugene Lih
- Center
for Biomaterials, Korea Institute of Science
and Technology, Seoul 02792, Republic of Korea
| | - Wooram Park
- Department
of Biomedical Science, College of Life Sciences, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Ki Wan Park
- Center
for Biomaterials, Korea Institute of Science
and Technology, Seoul 02792, Republic of Korea
- Department
of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Republic of Korea
| | - So Young Chun
- BioMedical
Research Institute, Kyungpook National University
Hospital, Daegu 41944, Republic of Korea
| | - Hyuncheol Kim
- Department
of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Republic of Korea
| | - Yoon Ki Joung
- Center
for Biomaterials, Korea Institute of Science
and Technology, Seoul 02792, Republic of Korea
| | - Tae Gyun Kwon
- Department
of Urology, School of Medicine, Kyungpook
National University, Daegu 37224, Republic of Korea
| | - Jeffrey A. Hubbell
- Institute
for Molecular Engineering, University of
Chicago, Chicago, Illinois 60637, United States
| | - Dong Keun Han
- Department
of Biomedical Science, College of Life Sciences, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, Gyeonggi 13488, Republic of Korea
| |
Collapse
|
50
|
Yang Y, Zhou X, Zhang M, Zhou Y, Wang B, Yuan C. Lateral wall osteotomy combined with embedded biodegradable implants for displaced intra-articular calcaneal fractures. J Orthop Surg Res 2019; 14:74. [PMID: 30841896 PMCID: PMC6402087 DOI: 10.1186/s13018-019-1111-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/21/2019] [Indexed: 11/10/2022] Open
Abstract
Background The extensile lateral approach (ELA) has been widely used to treat displaced intra-articular calcaneal fractures (DIACFs) and remains the gold standard procedure. Orthopedic surgeons are extremely concerned of the high rate of wound complications. This study intended to report a new surgical technique of the lateral wall osteotomy combined with an embedded biodegradable implant for treating DIACFs and assess clinical and radiological results. Methods From May 2013 to December 2015, a total of 17 patients with 19 calcaneal fractures underwent surgical treatment using our new technique. Radiographic images, computed tomography (CT) scans, and magnetic resonance (MR) images of the operative limb were obtained to assess fracture healing and biodegradable implant degradation. American Orthopaedic Foot and Ankle Society (AOFAS) ankle/hindfoot score at the last follow-up was obtained to assess functional result for all cases. Böhler’s and Gissane’s angles, width, and height of the injured calcaneus were analyzed using preoperative and last follow-up radiographic images. Results All radiological parameters were significantly improved at the last follow-up, with an increase of 15.58°, 8.38°, and 7.65 mm in Böhler’s angle, Gissane’s angle, and calcaneal height, respectively, and a decrease of 2.51 mm in calcaneal width (p < 0.05). Mean AOFAS score at the last follow-up was 84.37 ± 9.98, with 9, 6, and 4 feet, having excellent, good, and fair rates, respectively. None had nonunion, delayed union, or malunion after a mean follow-up of 34.69 ± 5.22 months. One superficial infection occurred 6 days post-surgery. Conclusions Osteotomy of the lateral wall of the calcaneus allows tension-free suturing and avoids damage to penetrating branches of the lateral calcaneal artery (LCA). Biodegradable implants are easy to reshape and do not require surgical removal. However, they should be limited to Sander’s type II and III fractures only. Level of evidence Level IV, case series without controls
Collapse
Affiliation(s)
- Yang Yang
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Xiaoxiao Zhou
- Department of Orthopedics, Zhoupu Hospital, Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Mengqin Zhang
- Intensive Care Unit, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Yichi Zhou
- Department of Orthopedics, CR & WISCO General Hospital, Wuhan, Hubei, China
| | - Bin Wang
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China.
| | - Chiting Yuan
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| |
Collapse
|