1
|
Liu S, Song L, Huang S, Liu Z, Xu Y, Wang Z, Qiu H, Wang J, Chen Z, Xiao Y, Wang H, Zhu X, Zhang K, Zhang X, Lin H. Hydroxyapatite microspheres encapsulated within hybrid hydrogel promote skin regeneration through the activation of Calcium Signaling and Motor Protein pathway. Bioact Mater 2025; 50:287-304. [PMID: 40292340 PMCID: PMC12022663 DOI: 10.1016/j.bioactmat.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/12/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Hydroxyapatite (HAp), traditionally recognized for its efficacy in bone regeneration, has rarely been explored for skin regeneration applications. This investigation explored HAp microspheres with distinct physicochemical properties tailored away from conventional bone regeneration parameters, and the capacity promoting skin regeneration and mitigating the aging process were investigated when encapsulated in hyaluronate hydrogels. By benchmarking against well-established dermal fillers like PMMA and PLLA, it was revealed the specific attributes of HAp that were conducive to skin regeneration, providing initial insights into the underlying mechanism. HAp enhanced the fibroblast functionality by triggering minimal adaptive immune responses and enhancing the Calcium Signaling and Motor Protein Signaling pathways. This modulation supported the production of normal collagen fibers, essential for ECM maturation and skin structural integrity. The significant ECM regeneration and remodeling capabilities exhibited by the HAp-encapsulated hybrid hydrogels suggested promising application in facial rejuvenation procedures, potentially making a breakthrough in aesthetic and reconstructive surgery.
Collapse
Affiliation(s)
- Shuo Liu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Lu Song
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Shuwen Huang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Zhanhong Liu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Yang Xu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Zhiyuan Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - He Qiu
- Department of Cosmetic and Plastic Surgery, West China School of Public, Health and West China Fourth Hospital, Sichuan University, Sichuan, Chengdu, China
| | - Jing Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Zhiru Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Yumei Xiao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
- Research Center for Material Genome Engineering, Sichuan University, Chengdu, 610064, China
| | - Hang Wang
- State Key Laboratory of Oral Diseases, Department of Cosmetic and Plastic, Surgery, Oral and Maxillofacial Surgery, National Clinical Research Center, for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu, 610064, China
- Research Center for Material Genome Engineering, Sichuan University, Chengdu, 610064, China
| | - Kai Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu, 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Hai Lin
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu, 610064, China
- Key Laboratory of Leather Chemistry and Engineering, Sichuan University, Ministry of Education, Chengdu, 610065, China
| |
Collapse
|
2
|
González A, Quibano-Ordoñez D, Ortega-Muñoz L, Moreno PA, Vélez-Varela PE. Genetic variants in the development of autoimmune complaints and capsular contracture in women with breast implants: A systematic review. JPRAS Open 2025; 44:529-541. [PMID: 40492145 PMCID: PMC12146490 DOI: 10.1016/j.jpra.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 04/06/2025] [Indexed: 06/11/2025] Open
Abstract
Introduction Breast implants were introduced in 1963 by Cronin and Gerow and are widely used for reconstructive and aesthetic purposes. However, their use has been controversial due to the potential risk of developing autoimmune diseases and surgical complications such as capsular contracture. Currently, no genetic markers have been identified that are definitively associated with these complications. Therefore, we conducted a systematic review to check for the presence of genetic variants that are related to complications such as capsular contracture and autoimmune diseases in women with breast implants. Materials and Methods A systematic review was conducted following the preferred reporting items for systematic reviews and meta-analyses guidelines. The search included databases such as Web of Science, PubMed, and Scopus, using terms such as "breast implants," "breast prostheses," "genes," "genetic markers," and "autoimmunity." The selected studies were cross-sectional, cohort, and case-control studies that assessed specific genetic variants in women with breast implants and their association with autoimmune diseases or capsular contracture. Results A total of 6 cross-sectional studies and 1 case-control study were included. The findings related to autoimmune diseases showed a high prevalence of the HLA-DQ10102 variant among women with myositis associated with breast implants compared to women with myositis without implants (81.8% vs. 31.6%; OR 9.8, 95% CI 1.77-96.79). Other studies identified genetic variants associated with capsular contracture, highlighting the expression of MMP, TIMP, TNF-α, and IL-8 genes in severe contractures. Conclusion Prolonged inflammation and specific genetic variants play a crucial role in the development of capsular contracture and autoimmune diseases in women with breast implants. Identifying these genetic markers could help improve the prediction and management of these complications. However, more longitudinal studies are needed to better understand the mechanisms and validate these findings.
Collapse
Affiliation(s)
- Alan González
- Esp. CareMe 360 Institute S.A.S. Barranquilla, COLOMBIA
| | - Daniela Quibano-Ordoñez
- Integrim S.A.S. Bogotá DC, COLOMBIA
- Department of Biology, FACNED, Universidad del Cauca, Popayán, COLOMBIA
| | | | - Pedro A. Moreno
- EISC, Faculty of Engineering. Universidad del Valle. Cali, COLOMBIA
| | | |
Collapse
|
3
|
Wiebe A, Hagemeister K, Fayyazi A, Apitzsch J, Lamadé W. Polyurethane-based tissue adhesive for sealing pancreatic anastomosis: a pig model. Surg Endosc 2025:10.1007/s00464-025-11802-5. [PMID: 40410619 DOI: 10.1007/s00464-025-11802-5] [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: 12/29/2024] [Accepted: 05/06/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND Pancreatic anastomosis is a critical step in partial pancreatoduodenectomy, as failure of the anastomosis with postoperative pancreatic fistula can lead to high morbidity and mortality. While various fibrin-based sealing agents have been tested, their effectiveness is limited due to rapid degradation by pancreatic secretions. This study aimed to evaluate the applicability and biocompatibility of a novel biodegradable, polyurethane-based adhesive (VIVO 120™) for intraoperative sealing of pancreatic anastomoses in a long-term survival pig model. METHODS An in vitro investigation of VIVO 120™ resistance to pancreatic enzymes over 21 days and an in vivo application for sealing a simplified pancreatogastrostomy were conducted. Following pancreatic tail resection, a simplified pancreatogastrostomy was performed in 12 pigs, sealing the anastomosis between the pancreatic body and the gastric wall with VIVO 120™. One pig underwent acute testing (1-day survival), and 11 pigs were included in a long-term survival study (14-day survival). Animals were divided into three groups based on adhesive application: Group 1 (intraluminal application, n = 4), Group 2 (extraluminal application, n = 3), and Group 3 (intraluminal application with pancreatic duct obstruction and superficial pressure injection into the pancreatic resection plane, n = 4). Clinical observations, laboratory diagnostics, necropsy, and histological analyses were performed. RESULTS VIVO 120™ exhibited high resistance to enzymatic degradation. Histology revealed no adhesive remnants in Group 1, whereas in Group 2, the adhesive was integrated into the surrounding connective tissue. Both groups showed signs of only very mild pancreatitis distant to the adhesive, likely resulting from surgical trauma, and no signs of pancreatic fistula, while Group 3 exhibited severe pancreatitis and extensive fibroblastic proliferation following pressure application. CONCLUSION VIVO 120™ is biocompatible and resistant to pancreatic secretions, suggesting its potential for effective sealing of pancreatic anastomoses. However, application with pressure into pancreatic tissue should be avoided to prevent severe pancreatitis and extensive fibroblastic proliferation.
Collapse
Affiliation(s)
- Anna Wiebe
- Department of Vascular Surgery, Klinikum Esslingen, Hirschlandstraße 97, 73730, Esslingen, Germany.
| | - Kerstin Hagemeister
- Universitätsklinikum Bonn, Haus für Experimentelle Therapie, Venusberg-Campus, Bonn, Germany
| | - Afshin Fayyazi
- SYNLAB Institute for Pathology and Molecular Pathology Pforzheim, Pforzheim, Germany
| | - Jonas Apitzsch
- Department of Radiology, Helios Klinikum Pforzheim, Pforzheim, Germany
| | - Wolfram Lamadé
- Department of General, Visceral and Vascular Surgery, Helios Klinikum Pforzheim, Pforzheim, Germany
| |
Collapse
|
4
|
Steverink JG, van Tol FR, Bruins S, Amponsah KB, Marvela J, Smorenburg AJ, Jonkman HR, Oosterman BJ, van Dijk MR, Malda J, Piluso S, Verlaan JJ. Testing a new sustained-release local anesthetic formulation specifically designed for spine surgery in a sheep model. Int J Pharm 2025; 678:125731. [PMID: 40389071 DOI: 10.1016/j.ijpharm.2025.125731] [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: 03/20/2025] [Revised: 05/12/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025]
Abstract
Instrumented spinal surgery is a frequently performed and painful intervention, with severe pain often lasting for three days. Opioids are the cornerstone of treatment of postoperative pain, but can induce severe side effects, dependence, and addiction. Novel local anesthetic applications, such as erector spinae plane blocks, and sustained release formulations have the potential to decrease opioid consumption without analgesic compromise but have limited efficacy beyond 24 h. A mismatch in duration of pain and duration of non-opioid analgesia is thus present in spine surgery. This study assessed the feasibility of a robust hydrogel for three days of sustained and stationary bupivacaine delivery, designed for co-implantation with pedicle screws, in a sheep model for spinal surgery. Fifteen sheep received six hydrogel rings loaded with bupivacaine mounted on pedicle screws (total dose 220 mg). Wound and systemic drug levels, hydrogel degradation and histological response were assessed. Moreover, in-vivo drug release was correlated to in-vitro release (IVIVC). Rings stayed in place after surgery and displayed a first-order release profile in-vivo over 72 h, with excellent IVIVC. Ring intactness after implantation did not affect release. Bupivacaine wound fluid levels exceeded plasma levels 2300-fold, and plasma bupivacaine levels were well below toxic thresholds through 168 h. Histological analysis of implant sites revealed a conventional foreign-body response that subsided during follow-up. Hydrogels degraded completely in 9 months. The present hydrogel has the potential to provide safe, localized, and sustained analgesia following instrumented spinal surgery.
Collapse
Affiliation(s)
- Jasper G Steverink
- Department of Orthopedic Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, Netherlands; SentryX BV, Yalelaan 54, 3584CM Utrecht, Netherlands.
| | - Floris R van Tol
- Department of Orthopedic Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, Netherlands; SentryX BV, Yalelaan 54, 3584CM Utrecht, Netherlands
| | | | | | | | | | | | | | - Marijke R van Dijk
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, Netherlands
| | - Jos Malda
- Department of Orthopedic Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584CM Utrecht, Netherlands
| | - Susanna Piluso
- Department of Orthopedic Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, Netherlands; SentryX BV, Yalelaan 54, 3584CM Utrecht, Netherlands
| | - Jorrit-Jan Verlaan
- Department of Orthopedic Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3584CX Utrecht, Netherlands; SentryX BV, Yalelaan 54, 3584CM Utrecht, Netherlands
| |
Collapse
|
5
|
Zhang C, Fu Z, Liu Q, Guo X, Li Z, Song W, Kong Y, Du J, Su Y, Yu B, Kong Y, Tian F, Fu X, Du X, Huang S. Bioprinted M2 macrophage-derived extracellular vesicle mimics attenuate foreign body reaction and enhance vascularized tissue regeneration. Biofabrication 2025; 17:035007. [PMID: 40328275 DOI: 10.1088/1758-5090/add49f] [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: 01/24/2025] [Accepted: 05/06/2025] [Indexed: 05/08/2025]
Abstract
Foreign body reaction (FBR) and insufficient vascularization greatly hinder the integration of 3D-bioprinted tissue substitutes with host tissues. Previous studies have shown that these problems are exacerbated by the stiffness of the 3D-bioprinted constructions, which is highly associated with the abnormal polarization of macrophages. Therefore, we developed an engineering strategy using membrane extrusion to prepare macrophage-derived extracellular vesicle mimics (EVMs). The EVMs derived from M1 and M2 macrophages (M1-EVMs and M2-EVMs) were rich in functional proteins. In the 2D environment, M1-EVMs promoted the fibrotic phenotype of fibroblasts, vascularization, and the M1 polarization of macrophages. In contrast, M2-EVMs effectively avoided the fibrotic trend, showed stronger angiogenic capabilities, and prevented excessive M1 polarization, demonstrating their potential to inhibit FBR and promote neovascularization. After bioprinting the EVMs loaded by gelatin-alginate bioink, the basic physical properties of the bioink were not significantly affected, and the biological functions of EVMs remain stable, indicating their potential as bioink additives. In the subcutaneous implantation model, unlike the FBR-aggravating effects of M1-EVMs, 3D-bioprinted M2-EVMs successfully reduced the immune response, prevented fibrous capsule formation, and increased vascular density. When applied to skin wound treatment, 3D-bioprinted M2-EVMs not only inhibited inflammatory levels but also exhibited pleiotropic pro-regenerative effects, effectively promoting vascularization, re-epithelialization, and appendage regeneration. As an innovative additive for bioinks, M2-EVMs present a promising approach to enhance the survival of bioengineered tissues and can further serve as a targeted drug loading system, promoting the development of regenerative medicine and improving clinical outcomes.
Collapse
Affiliation(s)
- Chao Zhang
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Ze Fu
- Chinese PLA Medical School, Beijing 100853, People's Republic of China
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Qinghua Liu
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Xu Guo
- College of Graduate, Tianjin Medical University, Tianjin 300203, People's Republic of China
| | - Zhao Li
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Wei Song
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Yi Kong
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Jinpeng Du
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Yanlin Su
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Bingyang Yu
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Yue Kong
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Feng Tian
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Xiaobing Fu
- School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- Chinese PLA Medical School, Beijing 100853, People's Republic of China
- College of Graduate, Tianjin Medical University, Tianjin 300203, People's Republic of China
| | - Xiaohui Du
- Chinese PLA Medical School, Beijing 100853, People's Republic of China
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| | - Sha Huang
- Medical Innovation Research Department, Research Center for Wound Repair and Tissue Regeneration, Chinese PLA General Hospital, Beijing 100048, People's Republic of China
- Chinese PLA Medical School, Beijing 100853, People's Republic of China
| |
Collapse
|
6
|
Huang W, Zong J, Li M, Li TF, Pan S, Xiao Z. Challenges and Opportunities: Nanomaterials in Epilepsy Diagnosis. ACS NANO 2025; 19:16224-16247. [PMID: 40266286 DOI: 10.1021/acsnano.5c01203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Epilepsy is a common neurological disorder characterized by a significant rate of disability. Accurate early diagnosis and precise localization of the epileptogenic zone are essential for timely intervention, seizure prevention, and personalized treatment. However, over 30% of patients with epilepsy exhibit negative results on electroencephalography and magnetic resonance imaging (MRI), which can lead to misdiagnosis and subsequent delays in treatment. Consequently, enhancing diagnostic methodologies is imperative for effective epilepsy management. The integration of nanomaterials with biomedicine has led to the development of diagnostic tools for epilepsy. Key advancements include nanomaterial-enhanced neural electrodes, contrast agents, and biochemical sensors. Nanomaterials improve the quality of electrophysiological signals and broaden the detection range of electrodes. In imaging, functionalized magnetic nanoparticles enhance MRI sensitivity, facilitating localization of the epileptogenic zone. NIR-II nanoprobes enable tracking of seizure-related biomarkers with deep tissue penetration. Furthermore, nanomaterials enhance the sensitivity of biochemical sensors for detecting epilepsy biomarkers, which is crucial for early detection. These advancements significantly increase diagnostic sensitivity and specificity. However, challenges remain, particularly regarding biosafety, quality control, and the scalability of fabrication processes. Overcoming these obstacles is essential for successful clinical translation. Artificial-intelligence-based big data analytics can facilitate the development of diagnostic tools by screening nanomaterials with specific properties. This approach may help to address current limitations and improve both effectiveness and safety. This review explores the application of nanomaterials in the diagnosis and detection of epilepsy, with the objective of inspiring innovative ideas and strategies to enhance diagnostic effectiveness.
Collapse
Affiliation(s)
- Wanbin Huang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiabin Zong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ming Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tong-Fei Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Songqing Pan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zheman Xiao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
7
|
Froom ZSCS, Callaghan NI, Davenport Huyer L. Cellular crosstalk in fibrosis: insights into macrophage and fibroblast dynamics. J Biol Chem 2025:110203. [PMID: 40334985 DOI: 10.1016/j.jbc.2025.110203] [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: 03/13/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Pathological fibrosis, the excessive deposition of extracellular matrix and tissue stiffening that causes progressive organ dysfunction, underlies diverse chronic diseases. The fibrotic microenvironment is driven by the dynamic microenvironmental interaction between various cell types; macrophages and fibroblasts play central roles in fibrotic disease initiation, maintenance, and progression. Macrophage functional plasticity to microenvironmental stimuli modulates fibroblast functionality by releasing pro-inflammatory cytokines, growth factors, and matrix remodeling enzymes that promote fibroblast proliferation, activation, and differentiation into myofibroblasts. Activated fibroblasts and myofibroblasts serve as the fibrotic effector cells, secreting extracellular matrix components and initiating microenvironmental contracture. Fibroblasts also modulate macrophage function through the release of their own pro-inflammatory cytokines and growth factors, creating bidirectional crosstalk that reinforces the chronic fibrotic cycle. The intricate interplay between macrophages and fibroblasts, including their secretomes and signaling interactions, leads to tissue damage and pathological loss of tissue function. In this review, we examine macrophage-fibroblast reciprocal dynamic interactions in pathological fibrotic conditions. We discuss the specific lineages and functionality of macrophages and fibroblasts implicated in fibrotic progression, with focus on their signal transduction pathways and secretory signalling that enables their pro-fibrotic behaviour. We then finish with a set of recommendations for future experimentation with the goal of developing a set of potential targets for anti-fibrotic therapeutic candidates. Understanding the cellular interactions between macrophages and fibroblasts provides valuable insights into potential therapeutic strategies to mitigate fibrotic disease progression.
Collapse
Affiliation(s)
- Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Neal I Callaghan
- Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada; Nova Scotia Health, Halifax, NS B3S 0H6, Canada.
| |
Collapse
|
8
|
Salehi Moghaddam A, Bahrami M, Sarikhani E, Tutar R, Ertas YN, Tamimi F, Hedayatnia A, Jugie C, Savoji H, Qureshi AT, Rizwan M, Maduka CV, Ashammakhi N. Engineering the Immune Response to Biomaterials. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414724. [PMID: 40232044 PMCID: PMC12097135 DOI: 10.1002/advs.202414724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/29/2025] [Indexed: 04/16/2025]
Abstract
Biomaterials are increasingly used as implants in the body, but they often elicit tissue reactions due to the immune system recognizing them as foreign bodies. These reactions typically involve the activation of innate immunity and the initiation of an inflammatory response, which can persist as chronic inflammation, causing implant failure. To reduce these risks, various strategies have been developed to modify the material composition, surface characteristics, or mechanical properties of biomaterials. Moreover, bioactive materials have emerged as a new class of biomaterials that can induce desirable tissue responses and form a strong bond between the implant and the host tissue. In recent years, different immunomodulatory strategies have been incorporated into biomaterials as drug delivery systems. Furthermore, more advanced molecule and cell-based immunomodulators have been developed and integrated with biomaterials. These emerging strategies will enable better control of the immune response to biomaterials and improve the function and longevity of implants and, ultimately, the outcome of biomaterial-based therapies.
Collapse
Affiliation(s)
- Abolfazl Salehi Moghaddam
- Department of BioengineeringP.C. Rossin College of Engineering & Applied ScienceLehigh UniversityBethlehemPA18015USA
| | - Mehran Bahrami
- Department of Mechanical Engineering & MechanicsLehigh UniversityBethlehemPA18015USA
| | - Einollah Sarikhani
- Department of Nano and Chemical EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Rumeysa Tutar
- Department of ChemistryFaculty of Engineering, Istanbul University‐CerrahpaşaIstanbul, Avcılar34320Turkey
| | - Yavuz Nuri Ertas
- Department of Biomedical EngineeringErciyes UniversityKayseri38039Turkey
- ERNAM – Nanotechnology Research and Application CenterErciyes UniversityKayseri38039Turkey
| | - Faleh Tamimi
- College of Dental MedicineQatar University HealthQatar UniversityP.O. Box 2713DohaQatar
| | - Ali Hedayatnia
- Azrieli Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Institute of Biomedical Engineering, Department of Pharmacology and PhysiologyFaculty of MedicineMontrealQuebecH3T 1J4Canada
- Montreal TransMedTech InstituteiTMTMontrealQuebecH3T 1J4Canada
| | - Clotilde Jugie
- Azrieli Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Montreal TransMedTech InstituteiTMTMontrealQuebecH3T 1J4Canada
| | - Houman Savoji
- Azrieli Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Institute of Biomedical Engineering, Department of Pharmacology and PhysiologyFaculty of MedicineMontrealQuebecH3T 1J4Canada
- Montreal TransMedTech InstituteiTMTMontrealQuebecH3T 1J4Canada
| | - Asma Talib Qureshi
- Department of Biomedical EngineeringMichigan Technological UniversityHoughtonMI49931USA
| | - Muhammad Rizwan
- Department of Biomedical EngineeringMichigan Technological UniversityHoughtonMI49931USA
- Health Research InstituteMichigan Technological UniversityHoughtonMI49931USA
| | - Chima V. Maduka
- BioFrontiers InstituteUniversity of ColoradoBoulderCO80303USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME)Colleges of Engineering and Human MedicineMichigan State UniversityEast LansingMI48824USA
- Department of BioengineeringSamueli School of EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
| |
Collapse
|
9
|
Wu X, Ye Y, Sun M, Mei Y, Ji B, Wang M, Song E. Recent Progress of Soft and Bioactive Materials in Flexible Bioelectronics. CYBORG AND BIONIC SYSTEMS 2025; 6:0192. [PMID: 40302943 PMCID: PMC12038164 DOI: 10.34133/cbsystems.0192] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/22/2024] [Accepted: 09/22/2024] [Indexed: 05/02/2025] Open
Abstract
Materials that establish functional, stable interfaces to targeted tissues for long-term monitoring/stimulation equipped with diagnostic/therapeutic capabilities represent breakthroughs in biomedical research and clinical medicine. A fundamental challenge is the mechanical and chemical mismatch between tissues and implants that ultimately results in device failure for corrosion by biofluids and associated foreign body response. Of particular interest is in the development of bioactive materials at the level of chemistry and mechanics for high-performance, minimally invasive function, simultaneously with tissue-like compliance and in vivo biocompatibility. This review summarizes the most recent progress for these purposes, with an emphasis on material properties such as foreign body response, on integration schemes with biological tissues, and on their use as bioelectronic platforms. The article begins with an overview of emerging classes of material platforms for bio-integration with proven utility in live animal models, as high performance and stable interfaces with different form factors. Subsequent sections review various classes of flexible, soft tissue-like materials, ranging from self-healing hydrogel/elastomer to bio-adhesive composites and to bioactive materials. Additional discussions highlight examples of active bioelectronic systems that support electrophysiological mapping, stimulation, and drug delivery as treatments of related diseases, at spatiotemporal resolutions that span from the cellular level to organ-scale dimension. Envisioned applications involve advanced implants for brain, cardiac, and other organ systems, with capabilities of bioactive materials that offer stability for human subjects and live animal models. Results will inspire continuing advancements in functions and benign interfaces to biological systems, thus yielding therapy and diagnostics for human healthcare.
Collapse
Affiliation(s)
- Xiaojun Wu
- Institute of Optoelectronics & Department of Materials Science, Shanghai Frontiers Science Research Base of Intelligent Optoelectronics and Perception, State Key Laboratory of Integrated Chips and Systems (SKLICS),
Fudan University, Shanghai 200438, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, State Key Laboratory of Molecular Engineering of Polymer,
Fudan University, Shanghai 200438, China
| | - Yuanming Ye
- Unmanned System Research Institute, National Key Laboratory of Unmanned Aerial Vehicle Technology, Integrated Research and Development Platform of Unmanned Aerial Vehicle Technology, Northwestern Polytechnical University, Xi’an 710072, China
- Queen Mary University of London Engineering School, Northwestern Polytechnical University, Xi’an 710072, China
| | - Mubai Sun
- Institute of Optoelectronics & Department of Materials Science, Shanghai Frontiers Science Research Base of Intelligent Optoelectronics and Perception, State Key Laboratory of Integrated Chips and Systems (SKLICS),
Fudan University, Shanghai 200438, China
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| | - Yongfeng Mei
- Institute of Optoelectronics & Department of Materials Science, Shanghai Frontiers Science Research Base of Intelligent Optoelectronics and Perception, State Key Laboratory of Integrated Chips and Systems (SKLICS),
Fudan University, Shanghai 200438, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, State Key Laboratory of Molecular Engineering of Polymer,
Fudan University, Shanghai 200438, China
- International Institute for Intelligent Nanorobots and Nanosystems,
Neuromodulation and Brain-machine-interface Centre, Fudan University, Shanghai 200438, China
- Yiwu Research Institute of Fudan University, Yiwu, Zhejiang 322000, China
| | - Bowen Ji
- Unmanned System Research Institute, National Key Laboratory of Unmanned Aerial Vehicle Technology, Integrated Research and Development Platform of Unmanned Aerial Vehicle Technology, Northwestern Polytechnical University, Xi’an 710072, China
| | - Ming Wang
- Institute of Optoelectronics & Department of Materials Science, Shanghai Frontiers Science Research Base of Intelligent Optoelectronics and Perception, State Key Laboratory of Integrated Chips and Systems (SKLICS),
Fudan University, Shanghai 200438, China
- Frontier Institute of Chip and System,
Fudan University, Shanghai 200433, China
| | - Enming Song
- Institute of Optoelectronics & Department of Materials Science, Shanghai Frontiers Science Research Base of Intelligent Optoelectronics and Perception, State Key Laboratory of Integrated Chips and Systems (SKLICS),
Fudan University, Shanghai 200438, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, State Key Laboratory of Molecular Engineering of Polymer,
Fudan University, Shanghai 200438, China
- International Institute for Intelligent Nanorobots and Nanosystems,
Neuromodulation and Brain-machine-interface Centre, Fudan University, Shanghai 200438, China
| |
Collapse
|
10
|
Lee SH, Yoo S, Kim SH, Kim YM, Han SI, Lee H. Nature-inspired surface modification strategies for implantable devices. Mater Today Bio 2025; 31:101615. [PMID: 40115053 PMCID: PMC11925587 DOI: 10.1016/j.mtbio.2025.101615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Medical and implantable devices are essential instruments in contemporary healthcare, improving patient quality of life and meeting diverse clinical requirements. However, ongoing problems such as bacterial colonization, biofilm development, foreign body responses, and insufficient device-tissue adhesion hinder the long-term effectiveness and stability of these devices. Traditional methods to alleviate these issues frequently prove inadequate, necessitating the investigation of nature-inspired alternatives. Biomimetic surfaces, inspired by the chemical and physical principles found in biological systems, present potential opportunities to address these challenges. Recent breakthroughs in manufacturing techniques, including lithography, vapor deposition, self-assembly, and three-dimensional printing, now permit precise control of surface properties at the micro- and nanoscale. Biomimetic coatings can diminish inflammation, prevent bacterial adherence, and enhance stable tissue integration by replicating the antifouling, antibacterial, and adhesive properties observed in creatures such as geckos, mussels, and biological membranes. This review emphasizes the cutting-edge advancements in biomimetic surfaces for medical and implantable devices, outlining their design methodologies, functional results, and prospective clinical applications. Biomimetic coatings, by integrating biological inspiration with advanced surface engineering, have the potential to revolutionize implantable medical devices, providing safer, more lasting, and more effective interfaces for prolonged patient benefit.
Collapse
Affiliation(s)
- Soo-Hwan Lee
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sungjae Yoo
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sung Hoon Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Young-Min Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Biomedical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Sang Ihn Han
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Biomedical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyojin Lee
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Biomedical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
- SKKU-KIST, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| |
Collapse
|
11
|
Hachim D, Hernández‐Cruz O, Foote JEJ, Wang R, Delahaye MW, Stuckey DJ, Feng Z, Wojciechowski JP, Salter LCB, Lin J, Harding SE, Stevens MM. Self-Doped and Biodegradable Glycosaminoglycan-PEDOT Conductive Hydrogels Facilitate Electrical Pacing of iPSC-Derived Cardiomyocytes. Adv Healthc Mater 2025; 14:e2403995. [PMID: 40018808 PMCID: PMC11973950 DOI: 10.1002/adhm.202403995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Conductive polymers hold promise in biomedical applications owing to their distinct conductivity characteristics and unique properties. However, incorporating these polymers into biomaterials poses challenges related to mechanical performance, electrical stability, and biodegradation. This study proposes an injectable hydrogel scaffold composed of a self-doped conductive polymer, constituted of a sulfated glycosaminoglycan (GAG) with side chains of PEDOT (poly 3,4-ethylenedioxythiophene). This brush copolymer is synthesized via oxidative polymerization from an EDOT monomer grafted onto the backbone of the sulfated GAG. The GAG backbone offers biodegradability, while sulfate groups act as acidic self-doping agents. Conductive hydrogels form through oxime crosslinking, initially existing as a liquid mixture that undergoes gelation within the tissue, allowing for injectability. The conductive hydrogels show tunable stiffness and gelation kinetics influenced by both concentration and pH, and exhibit adhesive properties. They showcase dual ionic and electronic conductivity, where sulfate groups in the GAG backbone act as doping moieties, enhancing conductivity and electrical stability. These properties of conductive hydrogels are associated with the facilitation of electrical pacing of iPSC-cardiomyocytes. Furthermore, hydrogels exhibit biodegradation and show evidence of biocompatibility, highlighting their potential for diverse biomedical applications.
Collapse
Affiliation(s)
- Daniel Hachim
- Department of MaterialsDepartment of Bioengineering and Institute of Biomedical EngineeringImperial College LondonExhibition RoadLondonSW7 2AZUK
- School of PharmacyFaculty of Chemistry and PharmacyPontifical Catholic University of ChileAV. VICUNA MACKENNA 4860Santiago7820436Chile
| | - Olivia Hernández‐Cruz
- Department of MaterialsDepartment of Bioengineering and Institute of Biomedical EngineeringImperial College LondonExhibition RoadLondonSW7 2AZUK
- National Heart and Lung InstituteImperial College LondonDu Cane RoadLondonW12 0NNUK
| | - James E. J. Foote
- Department of MaterialsDepartment of Bioengineering and Institute of Biomedical EngineeringImperial College LondonExhibition RoadLondonSW7 2AZUK
| | - Richard Wang
- Department of MaterialsDepartment of Bioengineering and Institute of Biomedical EngineeringImperial College LondonExhibition RoadLondonSW7 2AZUK
| | - Matthew W. Delahaye
- National Heart and Lung InstituteImperial College LondonDu Cane RoadLondonW12 0NNUK
| | - Daniel J. Stuckey
- Centre for Advanced Biomedical ImagingUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Zhiping Feng
- Centre for Advanced Biomedical ImagingUniversity College London72 Huntley StreetLondonWC1E 6DDUK
| | - Jonathan P. Wojciechowski
- Department of MaterialsDepartment of Bioengineering and Institute of Biomedical EngineeringImperial College LondonExhibition RoadLondonSW7 2AZUK
- Department of PhysiologyAnatomy and GeneticsDepartment of Engineering ScienceKavli Institute for Nanoscience DiscoveryUniversity of OxfordSherrington RoadOxfordOX1 3QUUK
| | - Luke C. B. Salter
- Department of MaterialsDepartment of Bioengineering and Institute of Biomedical EngineeringImperial College LondonExhibition RoadLondonSW7 2AZUK
| | - Junliang Lin
- Department of MaterialsDepartment of Bioengineering and Institute of Biomedical EngineeringImperial College LondonExhibition RoadLondonSW7 2AZUK
- Department of PhysiologyAnatomy and GeneticsDepartment of Engineering ScienceKavli Institute for Nanoscience DiscoveryUniversity of OxfordSherrington RoadOxfordOX1 3QUUK
| | - Sian E. Harding
- National Heart and Lung InstituteImperial College LondonDu Cane RoadLondonW12 0NNUK
| | - Molly M. Stevens
- Department of MaterialsDepartment of Bioengineering and Institute of Biomedical EngineeringImperial College LondonExhibition RoadLondonSW7 2AZUK
- Department of PhysiologyAnatomy and GeneticsDepartment of Engineering ScienceKavli Institute for Nanoscience DiscoveryUniversity of OxfordSherrington RoadOxfordOX1 3QUUK
| |
Collapse
|
12
|
Riew TR, Hwang JW, Kim TK, Kim YS. RNA Sequencing Revealed Distinct Expression Profiles and Temporal Expression Dynamics in Murine Model of Foreign Body Reaction. Exp Dermatol 2025; 34:e70104. [PMID: 40269512 DOI: 10.1111/exd.70104] [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: 02/14/2024] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025]
Abstract
Foreign body reaction (FBR) is an inflammatory and fibrotic reaction to degradation-resistant foreign materials characterised by the temporal cascade of cellular and molecular dynamics, which remains not fully elucidated. The aim of our study was to elucidate the temporal gene expression profiles of FBR. An FBR model was generated by implanting polycaprolactone into the abdominal subcutaneous layer of C57BL/6 mice. RNA sequencing was performed using established FBR tissues at various time points after implantation (FBR group; 2, 4, 8 and 12 weeks, n = 4 for each time points), and normal dorsal skin of mice as the control group (n = 3). We identified distinct gene expression profiles between the control group and the FBR group. Extracellular matrix (ECM), immune, and epigenetics-related genes were significantly enriched in the FBR group compared to normal skin. Within the FBR groups, expression profiles did not show definitive segregation across time points. We observed the highest expression of ECM-related genes (Adamts4, Col9a3, Col6a2, and Furin) and pathways in the 2-week samples, followed by a gradual down-regulation thereafter. In conclusion, our study elucidated distinct expression profiles of FBR in comparison to normal skin, as well as the temporal expression dynamics of FBR.
Collapse
Affiliation(s)
- Tae-Ryong Riew
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Sciences, Graduate School, the Catholic University of Korea, Seoul, Republic of Korea
| | - Ji-Won Hwang
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Sciences, Graduate School, the Catholic University of Korea, Seoul, Republic of Korea
| | - Tae Keun Kim
- Department of Dermatology, Bucheon St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon-Seob Kim
- Department of Dermatology, Bucheon St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
13
|
Durand M, Mathieu L, Venant J, Masquelet AC, Collombet JM. Engineering the bone reconstruction surgery: the case of the masquelet-induced membrane technique. Eur J Trauma Emerg Surg 2025; 51:138. [PMID: 40102268 PMCID: PMC11919993 DOI: 10.1007/s00068-025-02815-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/01/2025] [Indexed: 03/20/2025]
Abstract
The reconstruction of large bone defects remains challenging for orthopedic surgeons. Autologous bone grafts (ABGs) are the gold standard treatment for limited size defects, but larger bone defects (> 5 cm) require the use of more sophisticated techniques, such as the Masquelet technique. Over the last three decades, the Masquelet or induced membrane technique (IMT) has become increasingly popular as it does not require high-precision microsurgery skills and the time taken to achieve bone consolidation is independent of the length of the defect. IMT is a two-stage procedure. In the first stage, a polymethylmethacrylate (PMMA) cement spacer is implanted into the bone lesion and a physiological immune reaction initiates the formation of a fibrotic induced membrane (IM) with both angiogenic and osteogenic properties. The second stage, performed several weeks later, involves removal of the spacer followed by the implantation of a standard ABG in the preserved IM cavity for subsequent bone repair. In this extensive review, we explain how the success of this surgical procedure can be attributed to the synergy of four key components: the inducer (the PMMA cement), the recipient (the IM), the effector (the bone graft) and the modulator (the mechanical environment). Conversely, we then explain how each key component can contribute to the failure of such treatment. Finally, we discuss existing or emerging innovative and biotechnology-oriented strategies for optimizing surgical outcome with respect to the four components of IMT described above.
Collapse
Affiliation(s)
- Marjorie Durand
- Department of Medical and Surgical Assistance to the Armed Forces, French Armed Forces Biomedical Research Institute (IRBA), 1 Place du Général Valérie André, BP 40073, Brétigny sur Orge Cedex, 91222, France.
| | - Laurent Mathieu
- Department of Medical and Surgical Assistance to the Armed Forces, French Armed Forces Biomedical Research Institute (IRBA), 1 Place du Général Valérie André, BP 40073, Brétigny sur Orge Cedex, 91222, France
- Department of Orthopedic, Trauma and Reconstructive Surgery, Percy Military Hospital, 101 Avenue Henri Barbusse, Clamart, 92140, France
- Department of Hand and Upper Extremity Surgery, Edouard Herriot Hospital, 5 Place d'Arsonval, Lyon, 69003, France
- Department of Surgery, French Military Health Service Academy, 1 Place Alphonse Laveran, Paris, 75005, France
| | - Julien Venant
- Department of Medical and Surgical Assistance to the Armed Forces, French Armed Forces Biomedical Research Institute (IRBA), 1 Place du Général Valérie André, BP 40073, Brétigny sur Orge Cedex, 91222, France
| | | | - Jean-Marc Collombet
- Department of Medical and Surgical Assistance to the Armed Forces, French Armed Forces Biomedical Research Institute (IRBA), 1 Place du Général Valérie André, BP 40073, Brétigny sur Orge Cedex, 91222, France
| |
Collapse
|
14
|
Gao Y, Liang C, Yang B, Liao L, Su X. Application and Mechanism of Adipose Tissue-Derived Microvascular Fragments in Tissue Repair and Regeneration. Biomolecules 2025; 15:422. [PMID: 40149958 PMCID: PMC11939927 DOI: 10.3390/biom15030422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
One of the long-standing challenges in the field of tissue repair and regeneration is the rapid establishment of local microvascular circulation and restoration of perfusion at the site of defects or injuries. Recently, adipose tissue-derived microvascular fragments (ad-MVFs) have attracted increasing attention from researchers. Adipose tissue is rich in blood vessels, and significant progress has been made in the extraction and preservation techniques for microvascular fragments within it. Ad-MVFs promote tissue and organ repair and regeneration through three main mechanisms. First, they accelerate rapid and efficient vascularization at the injury site, enabling early vessel perfusion. Second, the stem cell components within ad-MVFs provide a rich source of cells for tissue and organ regeneration. Third, they play a role in immune regulation, facilitating integration with host tissues after implantation. The application methods of ad-MVFs are diverse. They can be directly implanted or pre-cultivated, facilitating their combination with various scaffolds and broadening their application scope. These properties have led to the wide use of ad-MVFs in tissue engineering, with promising prospects. This review demonstrates that ad-MVFs can serve as a reliable and highly feasible unit for tissue regeneration.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoxia Su
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine & Department of Pediatric, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; (Y.G.); (C.L.); (B.Y.); (L.L.)
| |
Collapse
|
15
|
Liu Z, Liu L, Liu J, Wu J, Tang R, Wolfram J. Electrospun meshes for abdominal wall hernia repair: Potential and challenges. Acta Biomater 2025; 195:52-72. [PMID: 39826853 DOI: 10.1016/j.actbio.2025.01.028] [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: 09/12/2024] [Revised: 12/11/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Surgical meshes are widely used in abdominal wall hernia repairs. However, consensus on mesh treatment remains elusive due to varying repair outcomes, especially with the introduction of new meshes, posing a substantial challenge for surgeons. Addressing these issues requires communicating the features of emerging candidates with a focus on clinical considerations. Electrospinning is a versatile technique for producing meshes with biomechanical architectures that closely mimic the extracellular matrix and enable incorporation of bioactive and therapeutic agents into the interconnective porous network, providing a favorable milieu for tissue integration and remodeling. Although this promising technique has drawn considerable interest in mesh fabrication and functionalization, currently developed electrospun meshes have limitations in meeting clinical requirements for hernia repair. This review summarizes the advantages and limitations of meshes prepared through electrospinning based on biomechanical, biocompatible, and bioactive properties/functions, offering interdisciplinary insights into challenges and future directions toward clinical mesh-aided hernia repair. STATEMENT OF SIGNIFICANCE: Consensus for hernia treatments using surgical meshes remains elusive based on varying repair outcomes, presenting significant challenges for researchers and surgeons. Differences in understanding mesh between specialists, particularly regarding material characteristics and clinical requirements, contribute to this issue. Electrospinning has been increasingly applied in mesh preparation through various approaches and strategies, aiming to improve abdominal wall hernia by restoring mechanical, morphological and functional integrity. However, there is no comprehensive overview of these emerging meshes regarding their features, functions, and clinical potentials, emphasizing the necessity of interdisciplinary discussions on this topic that build upon recent developments in electrospun mesh and provide insights from clinically practical prospectives.
Collapse
Affiliation(s)
- Zhengni Liu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia; Department of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, TongJi University, 150 Ji Mo Road, Shanghai, 200120, PR China.
| | - Lei Liu
- Department of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, TongJi University, 150 Ji Mo Road, Shanghai, 200120, PR China
| | - Jiajie Liu
- Department of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, TongJi University, 150 Ji Mo Road, Shanghai, 200120, PR China
| | - Jinglei Wu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, PR China
| | - Rui Tang
- Department of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, TongJi University, 150 Ji Mo Road, Shanghai, 200120, PR China
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia; School of Chemical Engineering, The University of Queensland, Brisbane, Queensland, 4072, Australia
| |
Collapse
|
16
|
Dalrymple AN, Jones ST, Fallon JB, Shepherd RK, Weber DJ. Overcoming failure: improving acceptance and success of implanted neural interfaces. Bioelectron Med 2025; 11:6. [PMID: 40083033 PMCID: PMC11907899 DOI: 10.1186/s42234-025-00168-7] [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: 12/18/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025] Open
Abstract
Implanted neural interfaces are electronic devices that stimulate or record from neurons with the purpose of improving the quality of life of people who suffer from neural injury or disease. Devices have been designed to interact with neurons throughout the body to treat a growing variety of conditions. The development and use of implanted neural interfaces is increasing steadily and has shown great success, with implants lasting for years to decades and improving the health and quality of life of many patient populations. Despite these successes, implanted neural interfaces face a multitude of challenges to remain effective for the lifetime of their users. The devices are comprised of several electronic and mechanical components that each may be susceptible to failure. Furthermore, implanted neural interfaces, like any foreign body, will evoke an immune response. The immune response will differ for implants in the central nervous system and peripheral nervous system, as well as over time, ultimately resulting in encapsulation of the device. This review describes the challenges faced by developers of neural interface systems, particularly devices already in use in humans. The mechanical and technological failure modes of each component of an implant system is described. The acute and chronic reactions to devices in the peripheral and central nervous system and how they affect system performance are depicted. Further, physical challenges such as micro and macro movements are reviewed. The clinical implications of device failures are summarized and a guide for determining the severity of complication was developed and provided. Common methods to diagnose and examine mechanical, technological, and biological failure modes at various stages of development and testing are outlined, with an emphasis on chronic in vivo characterization of implant systems. Finally, this review concludes with an overview of some of the innovative solutions developed to reduce or resolve the challenges faced by implanted neural interface systems.
Collapse
Affiliation(s)
- Ashley N Dalrymple
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
- Department of Physical Medicine and Rehabilitation, University of Utah, Salt Lake City, UT, USA.
- NERVES Lab, University of Utah, Salt Lake City, UT, USA.
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
- NeuroMechatronics Lab, Carnegie Mellon University, Pittsburgh, PA, USA.
| | - Sonny T Jones
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
- NERVES Lab, University of Utah, Salt Lake City, UT, USA
| | - James B Fallon
- Bionics Institute, St. Vincent's Hospital, Melbourne, VIC, Australia
- Medical Bionics Department, University of Melbourne, Melbourne, VIC, Australia
| | - Robert K Shepherd
- Bionics Institute, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Douglas J Weber
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
- NeuroMechatronics Lab, Carnegie Mellon University, Pittsburgh, PA, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
17
|
Cen C, Zhang Y, Cao Y, Hu C, Tang L, Liu C, Wang T, Peng W. Construction of a 3D Degradable PLLA/β-TCP/CS Scaffold for Establishing an Induced Membrane Inspired by the Modified Single-Stage Masquelet Technique. ACS Biomater Sci Eng 2025; 11:1629-1645. [PMID: 39943835 PMCID: PMC11900768 DOI: 10.1021/acsbiomaterials.4c01849] [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: 10/04/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 03/11/2025]
Abstract
Although the Masquelet-induced membrane technique (MIMT) is now employed worldwide for bone defects, it often needs to be repeated and autogenous bone graft. This study aims to investigate the theoretical feasibility of replacing PMMA (poly(methyl methacrylate)) bone cement with PLLA (poly-l-lactic acid)/β -TCP (beta-tricalcium phosphate)/CS (calcium sulfate) scaffold for single-stage bone defect reconstruction, which evoke the induced membrane (IM) formation in the early stage and directly acts as the implantation in the second stage to reconstruct the bone defect. We constructed a corn-like PLLA/β -TCP/CS scaffold by the fused deposition 3D printing method. The characterizations of the scaffolds were investigated systematically. The P/T15/S15 scaffolds (the PLLA/β -TCP/CS scaffold with a 15% mass fraction of β-TCP and 15% mass fraction of CS) were filled into the large-segmental radius bone defects of white rabbits to evoke the formation of IMs. HE (hematoxylin-eosin) and VG (van gieson) staining, along with immunofluorescent staining, were performed to analyze the architecture and cellularity, the expression of BMP-2 (bone morphogenetic protein-2), VEGF (vascular endothelial growth factor), and TGF-β1 (transforming growth factor-β1) was evaluated by IHC (immunohistochemistry) and WB (western-blot) respectively, the ALP (alkaline phosphatase) and ARS (alizarin red S) staining was applied to assess the osteogenic potential. The corn-like PLLA/β-TCP/CS scaffolds with excellent physicochemical properties are successfully constructed using the fused deposition 3D printing technique. The HE and VG staining, along with immunofluorescent staining, suggested that the P/T15/S15 scaffold effectively mediated the formation of IM after 6 weeks of placement. A significant presence of M2 macrophages was observed in IM. The results of IHC and WB demonstrated that the IMs derived from the P/T15/S15 scaffolds exhibited elevated levels of VEGF, BMP-2, and TGF-β1, all of which promote the osteogenic differentiation of BMSCs. The results of cellular immunofluorescence, flow cytometry, and WB indicate that P/T15/S15 regulates the phenotypic polarization of M0 macrophages toward the M2 phenotype via the PI3K/AKT/β-Catenin pathway. These findings suggest that the biodegradable PLLA/β-TCP/CS scaffold may serve as a viable alternative to PMMA bone cement for single-stage bone defect reconstruction, owing to its unique ability to stimulate IM formation and promote the polarization of macrophages toward the M2 phenotype. This work presents innovative materials and strategies for the management of bone defects.
Collapse
Affiliation(s)
- Chaode Cen
- School
of Clinical Medicine, The Guizhou Medical
University, Guiyang 550025,China
- Department
of Orthopedics, The Beijing Jishuitan Hospital
Guizhou Hospital, Guiyang 550014, China
| | - Yong Zhang
- Department
of gynaecology and obstetrics, Guiyang First
People’s Hospital, Guiyang 550005, China
| | - Yongfei Cao
- Department
of Orthopedics, The Beijing Jishuitan Hospital
Guizhou Hospital, Guiyang 550014, China
| | - Chaoran Hu
- Department
of Orthopedics, The Beijing Jishuitan Hospital
Guizhou Hospital, Guiyang 550014, China
| | - Lingli Tang
- Department
of Orthopedics, The Beijing Jishuitan Hospital
Guizhou Hospital, Guiyang 550014, China
| | - Chengwei Liu
- Department
of Orthopedics, The Beijing Jishuitan Hospital
Guizhou Hospital, Guiyang 550014, China
| | - Tao Wang
- School
of Clinical Medicine, The Guizhou Medical
University, Guiyang 550025,China
- Department
of Emergency Surgery, The Affiliated Hospital
of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Wuxun Peng
- School
of Clinical Medicine, The Guizhou Medical
University, Guiyang 550025,China
- Department
of Emergency Surgery, The Affiliated Hospital
of Guizhou Medical University, Guiyang, Guizhou 550004, China
| |
Collapse
|
18
|
Conner TS, Baaijens FPT, Bouten CVC, Angeloni L, Smits AIPM. A call for standardization: Evaluating different methodologies to induce in vitro foreign body giant cell formation for biomaterials research and design. Acta Biomater 2025; 194:20-37. [PMID: 39826854 DOI: 10.1016/j.actbio.2025.01.026] [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: 10/08/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Foreign body giant cells (FBGCs) are crucial in the foreign body reaction at the biomaterial-tissue interface, forming through the fusion of cells from the monocyte/macrophage lineage and performing functions such as material degradation and fibrous encapsulation. Yet, their presence and role in biomaterials research is only slowly unveiled. This review analyzed existing FBGC literature identified through a search string and sources from FBGC articles to evaluate the most commonly used methods and highlight the challenges in establishing a standardized protocol. Our findings revealed a fragmented research landscape marked by significant variability in in vitro culture conditions, i.e., cell origin and type, culture media and sera, fusion-inducing factors, seeding density, culture surface, and inconsistencies in the read-outs. This complicates efforts toward standardization and hampers cross-study comparisons. Based on these results, we highlight the need and propose guidelines for standardized culture protocols for FBGC research. Overall, this review aims to underscore the relevance of improving reproducibility and reliability in FBGC research, facilitating effective cross-study comparisons and advancing understanding of FBGC formation and function, ultimately contributing to designing more effective biomaterial-based therapies. STATEMENT OF SIGNIFICANCE: Foreign body giant cells (FBGCs) are crucial in the body's response to implanted biomaterials. Yet, current research addressing their role and impact is highly fragmented. This review comprehensively and systematically examines the diverse methodologies and definitions used in FBGC research and identifies critical gaps and inconsistencies hindering the reproducibility and comparison of findings. By advocating for standardized protocols, we aim to enhance the reliability and equivalence of research, thus providing a stronger foundation for understanding biomaterial-driven FBGC formation and function. Establishing such a framework will impact biomaterial-based therapies, supporting their effectiveness and safety in medical applications, and is thus of relevance for scientists, companies, and clinicians in the biomaterial and medical device communities.
Collapse
Affiliation(s)
- Thijs S Conner
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Frank P T Baaijens
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Livia Angeloni
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands; Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Rome, Italy
| | - Anthal I P M Smits
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands.
| |
Collapse
|
19
|
Miller A, Beck JP, White A, Agarwal J, Bachus KN, Jeyapalina S, Van Dyke M. Utilization of Bulk RNA Sequencing for the Evaluation of Keratin Nanomaterials as a Coating for Percutaneous Devices. J Biomed Mater Res B Appl Biomater 2025; 113:e35551. [PMID: 39962641 DOI: 10.1002/jbm.b.35551] [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: 09/12/2024] [Revised: 09/23/2024] [Accepted: 12/26/2024] [Indexed: 05/09/2025]
Abstract
Despite advances in the design and protocols for maintaining the skin/device interface around percutaneous devices (PDs), no current strategy ensures the permanent attachment of peri-implant epithelial tissue to the device surface. Based on preliminary data, we hypothesized that PDs coated with keratin nanomaterials, resembling the fingernail-nailbed interface, could provide a biochemically mediated surface that enhances epidermal cell adhesion and differentiation. To test this hypothesis, 15 Yucatan miniature pigs were each implanted with six percutaneous titanium devices, comprising three porous and three smooth devices, both with and without keratin coatings (Kerateine [iKNT] and Keratose [gKOS]). The pigs were sacrificed at 4, 8, and 16 weeks post-implantation. The devices and surrounding tissues were harvested and analyzed using histological and RNA sequencing techniques. Compared to smooth peri-implant tissue, porous peri-implant tissue showed a significant decrease in epithelial downgrowth, fibrous capsule thickness, and infection rates, alongside a significant upregulation of multiple immune marker genes, including IL12B. At the 16-week period, gKOS-coated surfaces demonstrated a more favorable wound healing response than iKTN-coated devices, with a reduction in granulation tissue area and a significant upregulation of several keratin genes related to differentiation. Among the combinations of surface types and coatings studied, the porous gKOS-coated device produced the most favorable wound healing response.
Collapse
Affiliation(s)
- Andrew Miller
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Division of Plastic Surgery, Department of Surgery, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - James Peter Beck
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Alexis White
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, Virginia, USA
| | - Jayant Agarwal
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Division of Plastic Surgery, Department of Surgery, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Kent N Bachus
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Sujee Jeyapalina
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
- Division of Plastic Surgery, Department of Surgery, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Mark Van Dyke
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
20
|
Gao L, Varley A, Gao H, Li B, Li X. Zwitterionic Hydrogels: From Synthetic Design to Biomedical Applications. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:3007-3026. [PMID: 39885654 DOI: 10.1021/acs.langmuir.4c04788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Zwitterionic hydrogels have emerged as a highly promising class of biomaterials, attracting considerable attention due to their unique properties and diverse biomedical applications. Zwitterionic moieties, with their balanced positive and negative charges, endow hydrogels with exceptional hydration, resistance to nonspecific protein adsorption, and low immunogenicity due to their distinctive molecular structure. These properties facilitate various biomedical applications, such as medical device coatings, tissue engineering, drug delivery, and biosensing. This review explores the structure-property relationships in zwitterionic hydrogels, highlighting recent advances in their design principles, synthesis methods, structural characteristics, and biomedical applications. To meet the evolving and growing demand for the biomedical field, this review examines current challenges and explores future research directions for optimizing the multifunctional properties of zwitterionic hydrogels. As promising candidates for advanced biomaterials, zwitterionic hydrogels are poised to address critical challenges in biomedical applications, paving the way for improved therapeutic outcomes and broader applicability in healthcare.
Collapse
Affiliation(s)
- Linran Gao
- State Key Laboratory of Separation Membranes and Membrane Processes, Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE), & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, China
| | - Andrew Varley
- RNA and Formulation Core, Michael Smith Laboratories, University of British Columbia, British Columbia, V6T 1Z4, Canada
| | - Hui Gao
- State Key Laboratory of Separation Membranes and Membrane Processes, Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE), & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, China
| | - Bowen Li
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada
| | - Xiaohui Li
- State Key Laboratory of Separation Membranes and Membrane Processes, Key Laboratory of Hollow Fiber Membrane Materials and Membrane Processes (MOE), & Tianjin Key Laboratory of Hollow Fiber Membrane Materials and Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, China
| |
Collapse
|
21
|
Park J, Kim D. Advanced Immunomodulatory Biomaterials for Therapeutic Applications. Adv Healthc Mater 2025; 14:e2304496. [PMID: 38716543 PMCID: PMC11834384 DOI: 10.1002/adhm.202304496] [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: 12/16/2023] [Revised: 04/15/2024] [Indexed: 05/22/2024]
Abstract
The multifaceted biological defense system modulating complex immune responses against pathogens and foreign materials plays a critical role in tissue homeostasis and disease progression. Recently developed biomaterials that can specifically regulate immune responses, nanoparticles, graphene, and functional hydrogels have contributed to the advancement of tissue engineering as well as disease treatment. The interaction between innate and adaptive immunity, collectively determining immune responses, can be regulated by mechanobiological recognition and adaptation of immune cells to the extracellular microenvironment. Therefore, applying immunomodulation to tissue regeneration and cancer therapy involves manipulating the properties of biomaterials by tailoring their composition in the context of the immune system. This review provides a comprehensive overview of how the physicochemical attributes of biomaterials determine immune responses, focusing on the physical properties that influence innate and adaptive immunity. This review also underscores the critical aspect of biomaterial-based immune engineering for the development of novel therapeutics and emphasizes the importance of understanding the biomaterials-mediated immunological mechanisms and their role in modulating the immune system.
Collapse
Affiliation(s)
- Ji‐Eun Park
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Dong‐Hwee Kim
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
- Department of Integrative Energy EngineeringCollege of EngineeringKorea UniversitySeoul02841Republic of Korea
- Biomedical Research CenterKorea Institute of Science and TechnologySeoul02792Republic of Korea
| |
Collapse
|
22
|
Zhao Y, Liu Y, Shan J, Xu X, Zhang C, Liu Z, Li X, Zhong Z, Gao Y, Ren K, Jiao D, Ren J, Wu P, Jiang Y, Han X. Anti-inflammatory coupled anti-angiogenic airway stent effectively suppresses tracheal in-stents restenosis. J Nanobiotechnology 2025; 23:59. [PMID: 39881307 PMCID: PMC11776288 DOI: 10.1186/s12951-024-03087-y] [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: 07/03/2024] [Accepted: 12/29/2024] [Indexed: 01/31/2025] Open
Abstract
Excessive vascularization during tracheal in-stent restenosis (TISR) is a significant but frequently overlooked issue. We developed an anti-inflammatory coupled anti-angiogenic airway stent (PAGL) incorporating anlotinib hydrochloride and silver nanoparticles using advanced electrospinning technology. PAGL exhibited hydrophobic surface properties, exceptional mechanical strength, and appropriate drug-release kinetics. Moreover, it demonstrated a remarkable eradication effect against methicillin-resistant Staphylococcus aureus. It also displayed anti-proliferative and anti-angiogenic properties on human umbilical vein endothelial cells and lung fibroblasts. PAGL was implanted into the tracheae of New Zealand rabbits to evaluate its efficacy in inhibiting bacterial infection, suppressing the inflammatory response, reducing angiogenesis, and attenuating excessive fibroblast activation. RNA sequencing analysis revealed a significant downregulation of genes associated with fibrosis, intimal hyperplasia, and cell migration following PAGL treatment. This study provides insight into the development of airway stents that target angiogenesis and inflammation to address problems associated with TISR effectively and have the potential for clinical translation.
Collapse
Affiliation(s)
- Yanan Zhao
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
- Interventional Institute of Zhengzhou University, Zhengzhou, 450001, China
| | - Yiming Liu
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
- Interventional Institute of Zhengzhou University, Zhengzhou, 450001, China
| | - Jiheng Shan
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
- Interventional Institute of Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaohong Xu
- State Key Laboratory of Antiviral Drugs, Henan Key Laboratory of Critical Care Medicine, Henan International Joint Laboratory of Infection and Immmunology, Department of Emergency Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, 450001, China
- School of Basic Medicine, Zhengzhou University, Zhengzhou, 450051, China
| | - Chengzhi Zhang
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
- Interventional Institute of Zhengzhou University, Zhengzhou, 450001, China
| | - Zaoqu Liu
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
- Interventional Institute of Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhaoqian Zhong
- State Key Laboratory of Antiviral Drugs, Henan Key Laboratory of Critical Care Medicine, Henan International Joint Laboratory of Infection and Immmunology, Department of Emergency Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, 450001, China
| | - Yanxia Gao
- State Key Laboratory of Antiviral Drugs, Henan Key Laboratory of Critical Care Medicine, Henan International Joint Laboratory of Infection and Immmunology, Department of Emergency Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, 450001, China
| | - Kewei Ren
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
- Interventional Institute of Zhengzhou University, Zhengzhou, 450001, China
| | - Dechao Jiao
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
- Interventional Institute of Zhengzhou University, Zhengzhou, 450001, China
| | - Jianzhuang Ren
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
- Interventional Institute of Zhengzhou University, Zhengzhou, 450001, China
| | - Ping Wu
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yong Jiang
- State Key Laboratory of Antiviral Drugs, Henan Key Laboratory of Critical Care Medicine, Henan International Joint Laboratory of Infection and Immmunology, Department of Emergency Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China.
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, 450001, China.
| | - Xinwei Han
- Department of Interventional Radiology, Key Laboratory of Interventional Radiology of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
23
|
Martí ML, Cano Aristizábal V, Motrich R, Valenti LE, Giacomelli CE. Defending Ti6Al4V against Biofilm Formation with Albumin Biofunctionalization. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:2089-2102. [PMID: 39812140 DOI: 10.1021/acs.langmuir.4c04867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Surface biofunctionalization with structurally perturbed albumin, as well as with other plasmatic proteins, inhibits the initial bacterial adhesion and biofilm formation, involved in numerous healthcare-associated infections. In fact, we have reported this protective effect with thermally treated plasmatic proteins, such as albumin and fibrinogen, adsorbed on flat silica surfaces. Here, we show that albumin biofunctionalization also works properly on flat Ti6Al4V substrates, which are widely used to fabricate medical devices. The protective effect is conserved even in biologically relevant fluids, containing other proteins that potentially adsorb onto and/or displace preadsorbed albumin from the biofunctionalized substrates. We further demonstrate that the presence of structurally perturbed albumin on the substrate does not trigger macrophage activation and the release of inflammatory mediators. Consequently, surface biofunctionalization with thermally perturbed albumin is a simple strategy to prepare antibacterial, nonimmunogenic medical devices.
Collapse
Affiliation(s)
- Ma Laura Martí
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Fisicoquímica, Ciudad Universitaria, X5000HUA Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Fisicoquímica de Córdoba (INFIQC), Ciudad Universitaria, X5000HUA Córdoba, Argentina
| | - Viviana Cano Aristizábal
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Fisicoquímica, Ciudad Universitaria, X5000HUA Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Fisicoquímica de Córdoba (INFIQC), Ciudad Universitaria, X5000HUA Córdoba, Argentina
| | - Rubén Motrich
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica clínica, Ciudad Universitaria, X5000HUA Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunologia (CIBICI), Ciudad Universitaria, X5000HUA Córdoba, Argentina
| | - Laura E Valenti
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Fisicoquímica, Ciudad Universitaria, X5000HUA Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Fisicoquímica de Córdoba (INFIQC), Ciudad Universitaria, X5000HUA Córdoba, Argentina
| | - Carla E Giacomelli
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Fisicoquímica, Ciudad Universitaria, X5000HUA Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Fisicoquímica de Córdoba (INFIQC), Ciudad Universitaria, X5000HUA Córdoba, Argentina
| |
Collapse
|
24
|
Robinson KJ, Voelcker NH, Thissen H. Clinical challenges and opportunities related to the biological responses experienced by indwelling and implantable bioelectronic medical devices. Acta Biomater 2025; 193:49-64. [PMID: 39675496 DOI: 10.1016/j.actbio.2024.12.037] [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: 07/30/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Implantable electrodes have been utilized for decades to stimulate, sense, or monitor a broad range of biological processes, with examples ranging from glucose monitoring devices to cochlear implants. While the underlying science related to the application of electrodes is a mature field, preclinical and clinical studies have demonstrated that there are still significant challenges in vivo associated with a lack of control over tissue-material interfacial interactions, especially over longer time frames. Herein we discuss the current challenges and opportunities for implantable electrodes and the associated bioelectronic interfaces across the clinical landscape with a focus on emerging technologies and the obstacles of biofouling, microbial colonization, and the foreign body response. Overcoming these challenges is predicted to open the door for a new generation of implantable medical devices and significant associated clinical impact. STATEMENT OF SIGNIFICANCE: Implantable electrodes have been utilised for decades to stimulate, sense, or monitor a broad range of biological processes, with examples ranging from glucose monitoring devices to cochlear implants. Next-generation bioelectronic implantable medical devices promise an explosion of new applications that have until this point in time been impossible to achieve. However, there are several persistent biological challenges hindering the realisation of these new applications. We present a clinical perspective on how these biological challenges have shaped the device market and clinical trial landscape. Specifically, we present statistical breakdowns of current device applications and discuss biofouling, the foreign body response, and microbial colonisation as the main factors that need to be addressed before a new generation of devices can be explored.
Collapse
Affiliation(s)
- Kye J Robinson
- CSIRO Manufacturing, Research Way, Clayton, Victoria 3168, Australia.
| | - Nicolas H Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia; Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
| | - Helmut Thissen
- CSIRO Manufacturing, Research Way, Clayton, Victoria 3168, Australia
| |
Collapse
|
25
|
Grande-Tovar CD, Castro Castro JI, Barba-Rosado LV, Zapata PA, Insuasty D, Valencia-Llano CH. Histology Assessment of Chitosan-Polyvinyl Alcohol Scaffolds Incorporated with CaO Nanoparticles. Molecules 2025; 30:276. [PMID: 39860146 PMCID: PMC11767540 DOI: 10.3390/molecules30020276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Scaffolds for regenerative therapy can be made from natural or synthetic polymers, each offering distinct benefits. Natural biopolymers like chitosan (CS) are biocompatible and biodegradable, supporting cell interactions, but lack mechanical strength. Synthetic polymers like polyvinyl alcohol (PVA) provide superior mechanical strength and cost efficiency but are not biodegradable or supportive of cell adhesion. Combining these polymers optimizes their advantages while adding metal oxide nanoparticles like calcium oxide (CaO NPs) enhances antimicrobial properties by damaging bacterial membranes. In this study, we obtained the formation of CaO NPs by calcinating eggshells, which were mixed in a polymeric network of CS and PVA to obtain four different membrane formulations for subdermal tissue regeneration. The spherical nanoparticles measured 13.43 ± 0.46 nm in size. Their incorporation into the membranes broadened the hydroxyl bands in the Fourier transform infrared (FTIR) analysis at 3331 cm⁻1. X-ray diffraction (XRD) analysis showed changes in the crystalline structure, with new diffraction peaks at 2θ values of 7.2° for formulations F2, F3, and F4, likely due to the increased amorphous nature and concentration of CaO NPs. Additionally, higher CaO NPs concentrations led to a reduction in thermal properties and crystallinity. Scanning electron microscopy (SEM) revealed a heterogeneous morphology with needle-like structures on the surface, resulting from the uniform dispersion of CaO NPs among the polymer chains and the solvent evaporation process. A histological examination of the implanted membranes after 60 days indicated their biocompatibility and biodegradability, facilitated by incorporating CaO NPs. During the degradation process, the material fragmented and was absorbed by inflammatory cells, which promoted the proliferation of collagen fibers and blood vessels. These findings highlight the potential of incorporating CaO NPs in soft tissue regeneration scaffolds.
Collapse
Affiliation(s)
- Carlos David Grande-Tovar
- Grupo de Investigación de Fotoquímica y Fotobiología, Universidad del Atlántico, Carrera 30 Número 8-49, Puerto Colombia 081008, Colombia;
| | - Jorge Ivan Castro Castro
- Tribology, Polymers, Powder Metallurgy and Solid Waste Transformations Research Group, Universidad del Valle, Calle 13 No. 100-00, Cali 760001, Colombia;
| | - Lemy Vanessa Barba-Rosado
- Grupo de Investigación de Fotoquímica y Fotobiología, Universidad del Atlántico, Carrera 30 Número 8-49, Puerto Colombia 081008, Colombia;
| | - Paula A. Zapata
- Grupo de Polímeros, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 9170020, Chile;
| | - Daniel Insuasty
- Departamento de Química y Biología, División de Ciencias Básicas, Universidad del Norte, Km 5 Vía Puerto Colombia, Barranquilla 081007, Colombia;
| | | |
Collapse
|
26
|
Zhou J, Zhang F, Tang Q, Zhu T, Ni Y, Wu Q, Liu Q, Zhu R, Wang T, Zhang Y, Zhang X, He H. Deoxygenated hydroxyapatite inhibits macrophage inflammation through fibronectin restricted adsorption. Acta Biomater 2025; 191:177-188. [PMID: 39577482 DOI: 10.1016/j.actbio.2024.11.026] [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: 07/16/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Macrophages can determine the ultimate outcome of the foreign body reaction (FBR). Although researchers confirmed that differences in the elemental composition of the implant interface can lead to varying levels of biological function, the mechanism underlying the polarization directions of macrophages induced by varying oxygen proportions remains unclear. This research presented the fabrication of a deoxygenated hydroxyapatite (dHAP) surface to investigate the impact of oxygen content on macrophage activation. The dHAP surface exhibited a pronounced inhibitory effect on the inflammatory activation of macrophages when compared to the HAP surface. Results from total internal reflection microscopy (TIRFM) and molecular dynamic (MD) simulation have revealed that the significant extracellular matrix adhesion protein, Fibronectin (Fn), showed a lower level of adsorption on dHAP surfaces. The Arg-Gly-Asp (RGD) structural domain showed a reduction in the exposure. The diminished adhesion capacity and impaired active site recognition ability of Fn resulted in lower activation of the integrin-focal adhesion kinase (FAK) pathway of macrophages on the dHAP surface, thereby suppressing the inflammation. In summary, this work explains the mechanism of the FBR impacted by the proportion of oxygen at the protein level. It also introduces a new approach to enhance the compatibility of biomaterials. STATEMENT OF SIGNIFICANCE: Macrophages are key in the foreign body response (FBR). Researches indicate that implant material's elemental interface content can regulate the functionality of biomaterials, but the mechanism of this regulation is unclear. To study the relationship between the elemental content at the interface and macrophages in the FBR, we prepared a deoxygenated hydroxyapatite (dHAP). Our results showed that the dHAP surface inhibited the adsorption behavior and changed the orientation of an ECM protein-fibronectin (Fn)-as well as the exposure of fewer active sites of the Arg-Gly-Asp (RGD) sequence, leading to less integrin activation. And then, the activation of the integrin- focal adhesion kinase (FAK) signaling pathway was reduced, leading to a greater activation of macrophages towards a pro-regenerative direction.
Collapse
Affiliation(s)
- Jingxuan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Fanyu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qinchao Tang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Taomin Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yueqi Ni
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qian Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qunli Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Runlin Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Tianman Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yufeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Medical Research Institute School of Medicine, Wuhan University, Wuhan 430071, China
| | - Xiaoxin Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Hong He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
27
|
Asadikorayem M, Weber P, Surman F, Puiggalí‐Jou A, Zenobi‐Wong M. Foreign Body Immune Response to Zwitterionic and Hyaluronic Acid Granular Hydrogels Made with Mechanical Fragmentation. Adv Healthc Mater 2025; 14:e2402890. [PMID: 39498680 PMCID: PMC11730820 DOI: 10.1002/adhm.202402890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/19/2024] [Indexed: 11/07/2024]
Abstract
Granular hydrogels have recently attracted the attention for diverse tissue engineering applications due to their versatility and modularity. Despite previous studies showing enhanced viability and metabolism of cells encapsulated in these hydrogels, the in vitro immune response and long-term fibrotic response of these scaffolds have not been well characterized. Here, bulk and granular hydrogels are studied based on synthetic zwitterionic (ZI) and natural polysaccharide hyaluronic acid (HA) made with mechanical fragmentation. In vitro, immunomodulatory studies show an increased stimulatory effect of HA granular hydrogels compared to bulk, while both bulk and granular ZI hydrogels do not induce an inflammatory response. Subcutaneous implantation in mice shows that both ZI and HA granular hydrogels resulted in less collagen capsule deposition around implants compared to bulk HA hydrogels 10 weeks after implantation. Moreover, the HA granular hydrogels are infiltrated by host cells, including macrophages and mature blood vessels, in a porosity-dependent manner. However, a large number of cells, including multinucleated giant cells as well as blood vessels, surround bulk and granular ZI hydrogels and are not able to infiltrate. Overall, this study provides new insights on the long-term stability and fibrotic response of granular hydrogels, paving the way for future studies and applications.
Collapse
Affiliation(s)
- Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and TechnologyETH ZürichOtto‐Stern‐Weg 7Zürich8093Switzerland
| | - Patrick Weber
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and TechnologyETH ZürichOtto‐Stern‐Weg 7Zürich8093Switzerland
| | - František Surman
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and TechnologyETH ZürichOtto‐Stern‐Weg 7Zürich8093Switzerland
| | - Anna Puiggalí‐Jou
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and TechnologyETH ZürichOtto‐Stern‐Weg 7Zürich8093Switzerland
| | - Marcy Zenobi‐Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and TechnologyETH ZürichOtto‐Stern‐Weg 7Zürich8093Switzerland
| |
Collapse
|
28
|
Tanaka ML, Saylor DM, Elder RM. Polymer-interface-tissue model to estimate leachable release from medical devices. MATHEMATICAL MEDICINE AND BIOLOGY : A JOURNAL OF THE IMA 2024; 41:382-403. [PMID: 39420619 DOI: 10.1093/imammb/dqae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
The ability to predict clinically relevant exposure to potentially hazardous compounds that can leach from polymeric components can help reduce testing needed to evaluate the biocompatibility of medical devices. In this manuscript, we compare two physics-based exposure models: 1) a simple, one-component model that assumes the only barrier to leaching is the migration of the compound through the polymer matrix and 2) a more clinically relevant, two-component model that also considers partitioning across the polymer-tissue interface and migration in the tissue away from the interface. Using data from the literature, the variation of the model parameters with key material properties were established, enabling the models to be applied to a wide range of combinations of leachable compound, polymer matrix and tissue type. Exposure predictions based on the models suggest that the models are indistinguishable over much of the range of clinically relevant scenarios. However, for systems with low partitioning and/or slow tissue diffusion, the two-component model predicted up to three orders of magnitude less mass release over the same time period. Thus, despite the added complexity, in some scenarios it can be beneficial to use the two-component model to provide more clinically relevant estimates of exposure to leachable substances from implanted devices.
Collapse
Affiliation(s)
- Martin L Tanaka
- College of Engineering and Technology, Western Carolina University, Cullowhee, NC 28723, USA
| | - David M Saylor
- Division of Biology, Chemistry, and Materials Science (DBCMS), Office of Science and Engineering Laboratories (OSEL), Center for Devices and Radiological Health (CDRH), US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Robert M Elder
- Division of Biology, Chemistry, and Materials Science (DBCMS), Office of Science and Engineering Laboratories (OSEL), Center for Devices and Radiological Health (CDRH), US Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
29
|
Wang J, Fan Z, Liu J, Liu K, Yan C, Ye X, Deng X. Influence of stent strut and its associated injury on thrombus formation: A dissipative particle dynamics study. J Theor Biol 2024; 595:111929. [PMID: 39197677 DOI: 10.1016/j.jtbi.2024.111929] [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: 03/22/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Vascular stent intervention is a pivotal treatment for coronary atherosclerosis, though in-stent thrombosis remains a significant postoperative complication with an unclear underlying mechanism. This study utilized dissipated particle dynamics analysis to investigate the impact of stent and its injury on platelet behavior. The findings suggest that thrombus formation upstream of the stent is mainly initiated by upstream arterial injury, which leads to increased platelet accumulation and activation in that area. While thrombosis downstream of the stent is more directly influenced by the stent itself. The morphology and size of in-stent thrombosis can vary significantly due to the different contributions of the stent and underlying injuries. Additionally, the volume of in-stent thrombosis is affected by the extent of the injury and the viscosity of platelets, showing a notable increase in volume with the lengthening of the injury area and rise in platelet viscosity. This study provides a novel theoretical framework for optimizing stent placement strategies and structural designs by examining the effects of stent struts and associated injuries on thrombus formation.
Collapse
Affiliation(s)
- Jian Wang
- School of Mechanical Engineering, Jiangsu University of Technology, Changzhou, Jiangsu 213001, China
| | - Zhenmin Fan
- School of Mechanical Engineering, Jiangsu University of Technology, Changzhou, Jiangsu 213001, China.
| | - Jiashuai Liu
- School of Mechanical Engineering, Jiangsu University of Technology, Changzhou, Jiangsu 213001, China
| | - Kailei Liu
- School of Mechanical Engineering, Jiangsu University of Technology, Changzhou, Jiangsu 213001, China
| | - ChaoJun Yan
- Cardiac Surgery Department, Southwest Hospital, Army Medical University, Chongqing 400038, China.
| | - Xia Ye
- School of Mechanical Engineering, Jiangsu University of Technology, Changzhou, Jiangsu 213001, China
| | - Xiaoyan Deng
- School of Mechanical Engineering, Jiangsu University of Technology, Changzhou, Jiangsu 213001, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
30
|
Nepon H, Julien C, Petrecca S, Kalashnikov N, Safran T, Murphy A, Dionisopoulos T, Davison P, Vorstenbosch J. The cellular and molecular properties of capsule surrounding silicone implants in humans vary uniquely according to the tissue type adjacent to the implant. J Biomed Mater Res A 2024; 112:2055-2070. [PMID: 38864257 DOI: 10.1002/jbm.a.37762] [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: 12/23/2023] [Revised: 04/15/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024]
Abstract
The foreign body reaction (FBR) to biomaterials results in fibrous encapsulation. Excessive capsule fibrosis (capsular contracture) is a major challenge to the long-term stability of implants. Clinical data suggests that the tissue type in contact with silicone breast implants alters susceptibility to developing capsular contracture; however, the tissue-specific inflammatory and fibrotic characteristics of capsule have not been well characterized at the cellular and molecular level. In this study, 60 breast implant capsule samples are collected from patients and stratified by the adjacent tissue type including subcutaneous tissue, glandular breast tissue, or muscle tissue. Capsule thickness, collagen organization, immune and fibrotic cellular populations, and expression of inflammatory and fibrotic markers is quantified with histological staining, immunohistochemistry, and real-time PCR. The findings suggest there are significant differences in M1-like macrophages, CD4+ T cells, CD26+ fibroblasts, and expression of IL-1β, IL-6, TGF-β, and collagen type 1 depending on the tissue type abutting the implant. Subglandular breast implant capsule displays a significant increase in inflammatory and fibrotic markers. These findings suggest that the tissue microenvironment contributes uniquely to the FBR. This data could provide new avenues for research and clinical applications to improve the site-specific biocompatibility and longevity of implantable devices.
Collapse
Affiliation(s)
- Hillary Nepon
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
- Division of Surgical and Interventional Sciences, McGill University, Montreal General Hospital, Quebec, Canada
| | - Cedric Julien
- McGill University Hospital Centre Research Institute, Montreal General Hospital, Quebec, Canada
| | - Sarah Petrecca
- Faculty of Medicine and Health Sciences, McGill University, Quebec, Canada
| | - Nikita Kalashnikov
- Division of Surgical and Interventional Sciences, McGill University, Montreal General Hospital, Quebec, Canada
- Faculty of Medicine and Health Sciences, McGill University, Quebec, Canada
| | - Tyler Safran
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
| | - Amanda Murphy
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
| | - Tassos Dionisopoulos
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
| | - Peter Davison
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
| | - Joshua Vorstenbosch
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, Quebec, Canada
- McGill University Hospital Centre Research Institute, Montreal General Hospital, Quebec, Canada
| |
Collapse
|
31
|
He S, Zhang Q, Jia J, Xia W, Chen S, Min F, Song Y, Yu Y, Li J, Li Z, Luo G. Stiffness and surface topology of silicone implants competitively mediate inflammatory responses of macrophages and foreign body response. Mater Today Bio 2024; 29:101304. [PMID: 39498150 PMCID: PMC11532915 DOI: 10.1016/j.mtbio.2024.101304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Adverse inflammatory responses, dominated by macrophages, that are induced by physical cues of silicone implants can heavily damage the life quality of patients via causing fibrosis and device failure. As stiffness and surface topology affect macrophages at the same time, the competition or partnership among physical cues against the regulation of macrophages is still ambiguous. Herein, a series of PDMS implants with different stiffness at ∼ MPa and surface topology at tens of micrometers were fabricated to investigate the relationship, the regulation rule, and the underlying mechanism of the two physical cues against the inflammatory responses of M1 macrophages. There is a competitive rule: surface topology could suppress the inflammatory responses of M1 macrophages in the soft group but did not have the same effect in the stiff group. Without surface topology, lower stiffness unexpectedly evoked stronger inflammatory responses of M1 macrophages. Implanting experiments also proved that the competitive state against mediating in vivo immune responses and the unexpected inflammatory responses. The reason is that stiffness could strongly up-regulate focal adhesion and activate the MAPK/NF-κB signaling axis to evoke inflammatory responses, which could shield the effect of surface topology. Therefore, for patient healthcare, it is crucial to prioritize stiffness while not surface topology at MPa levels to minimize adverse reactions.
Collapse
Affiliation(s)
- Sicen He
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Qingrong Zhang
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Jiezhi Jia
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Wei Xia
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Shengnan Chen
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing, 100190, PR China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Fanyi Min
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing, 100190, PR China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yanlin Song
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing, 100190, PR China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yunlong Yu
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Jiangfeng Li
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Zheng Li
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Gaoxing Luo
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| |
Collapse
|
32
|
Chen S, Du T, Zhang H, Zhang Y, Qiao A. Advances in studies on tracheal stent design addressing the related complications. Mater Today Bio 2024; 29:101263. [PMID: 39399242 PMCID: PMC11467681 DOI: 10.1016/j.mtbio.2024.101263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/09/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024] Open
Abstract
Tracheal stents can be used to quickly reconstruct the airway and relieve symptoms of dyspnea in patients with tracheal stenosis. However, existing tracheal stents lead to complications such as granulation tissue formation, difficulty in removal, persistent growth of malignant tumors, stent migration, and mucus plugging. In this article, we reviewed the main methods used to reduce complications associated with tracheal stent design. Drug-eluting stents can inhibit granulation tissue formation and prevent infection and local chemotherapy. The biodegradable stent can support the trachea for some time, maintain tracheal patency, and degrade gradually, which avoids removing or replacing the stent. Radioactive stents loaded with I125 have good potential for inhibiting the persistent growth of malignant tumors. Three-dimensional printing technology enables the manufacturing of patient-specific stents, which increases the degree of matching between the complex tracheal anatomy and the stent, thus providing a new solution for stent migration caused by structural mismatch. Minimizing the barrier of the stent to mucociliary clearance, providing an anti-fouling coating, and culturing respiratory epithelial cells on the surface of the stent are the main methods used to reduce mucus plugging. We also proposed future research directions for tracheal stents to guide the design and manufacture of ideal tracheal stents.
Collapse
Affiliation(s)
- Shiliang Chen
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
- Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing University of Technology, Beijing, China
| | - Tianming Du
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
- Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing University of Technology, Beijing, China
| | - Hanbing Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
- Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing University of Technology, Beijing, China
| | - Yanping Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
- Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing University of Technology, Beijing, China
| | - Aike Qiao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
- Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing University of Technology, Beijing, China
| |
Collapse
|
33
|
Schoberleitner I, Faserl K, Lackner M, Coraça-Huber DC, Augustin A, Imsirovic A, Sigl S, Wolfram D. Unraveling the Immune Web: Advances in SMI Capsular Fibrosis from Molecular Insights to Preclinical Breakthroughs. Biomolecules 2024; 14:1433. [PMID: 39595609 PMCID: PMC11592141 DOI: 10.3390/biom14111433] [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: 10/07/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Breast implant surgery has evolved significantly, yet challenges such as capsular contracture remain a persistent concern. This review presents an in-depth analysis of recent advancements in understanding the immune mechanisms and clinical implications associated with silicone mammary implants (SMIs). The article systematically examines the complex interplay between immune responses and capsular fibrosis, emphasizing the pathophysiological mechanisms of inflammation in the etiology of this fibrotic response. It discusses innovations in biomaterial science, including the development of novel anti-biofilm coatings and immunomodulatory surfaces designed to enhance implant integration and minimize complications. Emphasis is placed on personalized risk assessment strategies, leveraging molecular insights to tailor interventions and improve patient outcomes. Emerging therapeutic targets, advancements in surgical techniques, and the refinement of post-operative care are also explored. Despite notable progress, challenges such as the variability in immune responses, the long-term efficacy of new interventions, and ethical considerations remain. Future research directions are identified, focusing on personalized medicine, advanced biomaterials, and bridging preclinical findings with clinical applications. As we advance from bench to bedside, this review illuminates the path forward, where interdisciplinary collaboration and continued inquiry weave together to enhance the art and science of breast implant surgery, transforming patient care into a realm of precision and excellence.
Collapse
Affiliation(s)
- Ines Schoberleitner
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Müllerstraße 44, 6020 Innsbruck, Austria
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Klaus Faserl
- Protein Core Facility, Institute of Medical Chemistry, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria
| | - Débora C. Coraça-Huber
- BIOFILM Lab, Department of Orthopedics and Traumatology, Medical University of Innsbruck, Müllerstraße 44, 6020 Innsbruck, Austria
| | - Angela Augustin
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Anja Imsirovic
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Stephan Sigl
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Dolores Wolfram
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| |
Collapse
|
34
|
Peltokallio NMM, Ajdary R, Reyes G, Kankuri E, Junnila JJT, Kuure S, Meller AS, Kuula J, Raussi-Lehto E, Sariola H, Laitinen-Vapaavuori OM, Rojas OJ. Comparative In Vivo Biocompatibility of Cellulose-Derived and Synthetic Meshes in Subcutaneous Transplantation Models. Biomacromolecules 2024; 25:7298-7310. [PMID: 39376005 PMCID: PMC11558565 DOI: 10.1021/acs.biomac.4c00984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024]
Abstract
Despite the increasing interest in cellulose-derived materials in biomedical research, there remains a significant gap in comprehensive in vivo analyses of cellulosic materials obtained from various sources and processing methods. To explore durable alternatives to synthetic medical meshes, we evaluated the in vivo biocompatibility of bacterial nanocellulose, regenerated cellulose, and cellulose nanofibrils in a subcutaneous transplantation model, alongside incumbent polypropylene and polydioxanone. Notably, this study demonstrates the in vivo biocompatibility of regenerated cellulose obtained through alkali dissolution and subsequent regeneration. All cellulose-derived implants triggered the expected foreign body response in the host tissue, characterized predominantly by macrophages and foreign body giant cells. Porous materials promoted cell ingrowth and biointegration. Our results highlight the potential of bacterial nanocellulose and regenerated cellulose as safe alternatives to commercial polypropylene meshes. However, the in vivo fragmentation observed for cellulose nanofibril meshes suggests the need for measures to optimize their processing and preparation.
Collapse
Affiliation(s)
- Nina M. M. Peltokallio
- Department
of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Viikintie 49, FI-00014 Helsinki University, Finland
| | - Rubina Ajdary
- Biobased
Colloids and Materials, Department of Bioproducts and Biosystems,
School of Chemical Engineering, Aalto University, P.O. Box 16300, FI-00076 Aalto, Espoo,Finland
| | - Guillermo Reyes
- Biobased
Colloids and Materials, Department of Bioproducts and Biosystems,
School of Chemical Engineering, Aalto University, P.O. Box 16300, FI-00076 Aalto, Espoo,Finland
| | - Esko Kankuri
- Department
of Pharmacology, Faculty of Medicine, University
of Helsinki, P.O. Box 29, Helsinki 00014, Finland
| | | | - Satu Kuure
- GM
unit, Helsinki Institute of Life Science/STEMM, Research Program′s
Unit, Faculty of Medicine, University of
Helsinki, P.O. Box 63, Helsinki 00014, Finland
| | - Anna S. Meller
- Laboratory
Animal Centre, HiLIFE, University of Helsinki, P.O. Box 29, Helsinki 00014, Finland
| | - Jani Kuula
- Department
of Neuroscience and Biomedical Engineering, School of Science, Aalto University, P.O.
Box 16300, FI-00076 Aalto, Espoo, Finland
| | - Eija Raussi-Lehto
- Department
of Neuroscience and Biomedical Engineering, School of Science, Aalto University, P.O.
Box 16300, FI-00076 Aalto, Espoo, Finland
- Customer-oriented
Wellbeing and Health Services, Metropolia
University of Applied Sciences, PL 4000, FI-00079 Metropolia, Helsinki,Finland
| | - Hannu Sariola
- Department
of Pathology, Faculty of Medicine, University
of Helsinki, P.O. Box 63, Helsinki 00014, Finland
| | - Outi M. Laitinen-Vapaavuori
- Department
of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Viikintie 49, FI-00014 Helsinki University, Finland
| | - Orlando J. Rojas
- Biobased
Colloids and Materials, Department of Bioproducts and Biosystems,
School of Chemical Engineering, Aalto University, P.O. Box 16300, FI-00076 Aalto, Espoo,Finland
- Bioproducts
Institute, Department of Chemical and Biological Engineering, The University of British Columbia, 2360 East Mall, Vancouver, BC V6T 1Z3, Canada
- Department of Wood
Science, University
of British Columbia, 2385 East Mall, Vancouver, BC V6T 1Z4, Canada
- Department of Chemistry, University of
British Columbia, 2036
Main Mall, Vancouver, BC V6T 1Z1, Canada
| |
Collapse
|
35
|
Guan X, Yao H, Wu J. Photocrosslinkable hydrogel of ibuprofen-chitosan methacrylate modulates inflammatory response. J Biomed Mater Res A 2024; 112:2001-2017. [PMID: 38837524 DOI: 10.1002/jbm.a.37758] [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: 03/30/2024] [Revised: 05/20/2024] [Accepted: 05/25/2024] [Indexed: 06/07/2024]
Abstract
Methacrylated biopolymers are unique and attractive in preparing photocrosslinkable hydrogels in biomedical applications. Here we report a novel chitosan (CS) derivative-based injectable hydrogel with anti-inflammatory capacity via methacrylation modification. First, ibuprofen (IBU) was conjugated to the backbone of CS by carbodiimide chemistry to obtain IBU-CS conjugate, which converts water-insoluble unmodified CS into water-soluble IBU-CS conjugate. The IBU-CS conjugate did not precipitate at the pH of 7, which was beneficial to subsequent chemical modification with methacrylic anhydride to prepare IBU-CS methacrylate (IBU-CS-MA) with significantly higher methacrylation substitution. Photocrosslinkable in situ gel formation of injectable IBU-CS-MA hydrogel was verified using lithium phenyl-2,4,6-trimethylbenzoylphosphinate (LAP) initiator under visible light. The IBU-CS-MA hydrogel showed good cytocompatibility as revealed by encapsulating and in vitro culturing murine fibroblasts within hydrogels. It promoted macrophage polarization toward M2 phenotype, as well as downregulated pro-inflammatory gene expression and upregulated anti-inflammatory gene expression of macrophages. The hydrogel also significantly reduced the reactive oxygen specifies (ROS) and nitrogen oxide (NO) produced by lipopolysaccharides (LPS)-stimulated macrophages. Upon subcutaneous implantation in a rat model, it significantly mitigated inflammatory responses as shown by significantly lower inflammatory cell density, less cell infiltration, and much thinner fibrous capsule compared with CS methacrylate (CS-MA) hydrogel. This study suggests that IBU-CS conjugate represents a feasible strategy for preparing CS-based methacrylate hydrogels for biomedical applications.
Collapse
Affiliation(s)
- Xiangheng Guan
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Department of Biomedical Engineering, Donghua University, Shanghai, People's Republic of China
| | - Haochen Yao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, People's Republic of China
| | - Jinglei Wu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Department of Biomedical Engineering, Donghua University, Shanghai, People's Republic of China
| |
Collapse
|
36
|
Guan X, Wang XG, Sun B, Wang H, El-Newehy M, Abdulhameed MM, Mo X, Feng B, Wu J. A photocrosslinkable and anti-inflammatory hydrogel of loxoprofen-conjugated chitosan methacrylate. J Mater Chem B 2024. [PMID: 39470461 DOI: 10.1039/d4tb01956c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Polymer-drug conjugates are widely used for drug delivery. Herein, we report an injectable hydrogel for local delivery of nonsteroidal anti-inflammatory drugs (NSAIDs) using chitosan (CS) as a carrier polymer. Loxoprofen (LOX) was conjugated to the backbone of CS via carbodiimide chemistry to obtain the LOX-CS conjugate. This conjugation transformed the water-insoluble unmodified CS into the water-soluble LOX-CS conjugate. In particular, the LOX-CS conjugate did not precipitate at pH 7, allowing smooth subsequent chemical modification with methacrylic anhydride (MA) to synthesize LOX-CS methacrylate (LOX-CS-MA) with significantly higher methacrylation substitution. The LOX-CS-MA was capable of in situ gel formation under visible light irradiation in the presence of a benzoin-2,4,6-trimethylbenzoylphosphinate lithium (LAP) photoinitiator. Our results show that the LOX-CS-MA hydrogel exhibited good cytocompatibility and blood compatibility. It promoted M2 polarization, inhibited pro-inflammatory gene expression, and upregulated anti-inflammatory gene expression of macrophages. Furthermore, the LOX-CS-MA hydrogel significantly reduced reactive oxygen species (ROS) and nitric oxide (NO) produced by lipopolysaccharide (LPS)-stimulated macrophages. A subcutaneous implanted LOX-CS-MA hydrogel in a rat model revealed significantly reduced inflammatory cell density, decreased cell infiltration, and a much thinner fibrous capsule compared to the CS methacrylate (CS-MA) hydrogel, thus markedly alleviating the inflammatory response. This study highlights the feasibility of CS-drug conjugates in preparing CS-based methacrylate hydrogels for sustained drug release.
Collapse
Affiliation(s)
- Xiangheng Guan
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Department of Biomedical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Xin-Gang Wang
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China.
| | - Binbin Sun
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Department of Biomedical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Hongsheng Wang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Department of Biomedical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Mohamed El-Newehy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Meera Moydeen Abdulhameed
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Xiumei Mo
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Department of Biomedical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Bei Feng
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China.
| | - Jinglei Wu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Department of Biomedical Engineering, Donghua University, Shanghai 201620, P. R. China.
| |
Collapse
|
37
|
Cai F, Jiang B, He F. Formation and biological activities of foreign body giant cells in response to biomaterials. Acta Biomater 2024; 188:1-26. [PMID: 39245307 DOI: 10.1016/j.actbio.2024.08.034] [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: 03/09/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 09/10/2024]
Abstract
The integration of biomaterials in medical applications triggers the foreign body response (FBR), a multi-stage immune reaction characterized by the formation of foreign body giant cells (FBGCs). Originating from the fusion of monocyte/macrophage lineage cells, FBGCs are pivotal participants during tissue-material interactions. This review provides an in-depth examination of the molecular processes during FBGC formation, highlighting signaling pathways and fusion mediators in response to both exogenous and endogenous stimuli. Moreover, a wide range of material-specific characteristics, such as surface chemical and physical properties, has been proven to influence the fusion of macrophages into FBGCs. Multifaceted biological activities of FBGCs are also explored, with emphasis on their phagocytic capabilities and extracellular secretory functions, which profoundly affect the vascularization, degradation, and encapsulation of the biomaterials. This review further elucidates the heterogeneity of FBGCs and their diverse roles during FBR, as demonstrated by their distinct behaviors in response to different materials. By presenting a comprehensive understanding of FBGCs, this review intends to provide strategies and insights into optimizing biocompatibility and the therapeutic potential of biomaterials for enhanced stability and efficacy in clinical applications. STATEMENT OF SIGNIFICANCE: As a hallmark of the foreign body response (FBR), foreign body giant cells (FBGCs) significantly impact the success of implantable biomaterials, potentially leading to complications such as chronic inflammation, fibrosis, and device failure. Understanding the role of FBGCs and modulating their responses are vital for successful material applications. This review provides a comprehensive overview of the molecules and signaling pathways guiding macrophage fusion into FBGCs. By elucidating the physical and chemical properties of materials inducing distinct levels of FBGCs, potential strategies of materials in modulating FBGC formation are investigated. Additionally, the biological activities of FBGCs and their heterogeneity in responses to different material categories in vivo are highlighted in this review, offering crucial insights for improving the biocompatibility and efficacy of biomaterials.
Collapse
Affiliation(s)
- Fangyuan Cai
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Bulin Jiang
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Fuming He
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
38
|
Amani H, Alipour M, Shahriari E, Taboas JM. Immunomodulatory Biomaterials: Tailoring Surface Properties to Mitigate Foreign Body Reaction and Enhance Tissue Regeneration. Adv Healthc Mater 2024:e2401253. [PMID: 39370571 DOI: 10.1002/adhm.202401253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/28/2024] [Indexed: 10/08/2024]
Abstract
The immune cells have demonstrated the ability to promote tissue repair by removing debris, breaking down the extracellular matrix, and regulating cytokine secretion profile. If the behavior of immune cells is not well directed, chronic inflammation and foreign body reaction (FBR) will lead to scar formation and loss of biomaterial functionality. The immunologic response toward tissue repair or chronic inflammation after injury and implantation can be modulated by manipulating the surface properties of biomaterials. Tailoring surface properties of biomaterials enables the regulation of immune cell fate such as adhesion, proliferation, recruitment, polarization, and cytokine secretion profile. This review begins with an overview of the role of immune cells in tissue healing and their interactions with biomaterials. It then discusses how the surface properties of biomaterials influence immune cell behavior. The core focus is reviewing surface modification methods to create innovative materials that reduce foreign body reactions and enhance tissue repair and regeneration by modulating immune cell activities. The review concludes with insights into future advancements in surface modification techniques and the associated challenges.
Collapse
Affiliation(s)
- Hamed Amani
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mahdieh Alipour
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Elahe Shahriari
- Department of Physiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Juan M Taboas
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| |
Collapse
|
39
|
Roosa CA, Lempke SL, Hannan RT, Nicklow E, Sturek JM, Ewald SE, Griffin D. Conjugation of IL-33 to Microporous Annealed Particle Scaffolds Enhances Type 2-Like Immune Responses In Vitro and In Vivo. Adv Healthc Mater 2024; 13:e2400249. [PMID: 38648258 PMCID: PMC11461124 DOI: 10.1002/adhm.202400249] [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: 02/09/2024] [Revised: 04/11/2024] [Indexed: 04/25/2024]
Abstract
The inflammatory foreign body response (FBR) is the main driver of biomaterial implant failure. Current strategies to mitigate the onset of a FBR include modification of the implant surface, release of anti-inflammatory drugs, and cell-scale implant porosity. The microporous annealed particle (MAP) scaffold platform is an injectable, porous biomaterial composed of individual microgels, which are annealed in situ to provide a structurally stable scaffold with cell-scale microporosity. MAP scaffold does not induce a discernible foreign body response in vivo and, therefore, can be used a "blank canvas" for biomaterial-mediated immunomodulation. Damage associated molecular patterns (DAMPs), such as IL-33, are potent regulators of type 2 immunity that play an important role in tissue repair. In this manuscript, IL-33 is conjugated to the microgel building-blocks of MAP scaffold to generate a bioactive material (IL33-MAP) capable of stimulating macrophages in vitro via a ST-2 receptor dependent pathway and modulating immune cell recruitment to the implant site in vivo, which indicates an upregulation of a type 2-like immune response and downregulation of a type 1-like immune response.
Collapse
Affiliation(s)
- Colleen A. Roosa
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd, Charlottesville, Virginia 22903, USA
| | - Samantha L. Lempke
- Department of Microbiology, Immunology, and Cancer Biology, Beirne B. Carter Immunology Center, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, Virginia 22903, USA
| | - Riley T. Hannan
- Department of Medicine, Pulmonary and Critical Care, University of Virginia, 1221 Lee St, Charlottesville, Virginia 22903, USA
| | - Ethan Nicklow
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd, Charlottesville, Virginia 22903, USA
| | - Jeffrey M. Sturek
- Department of Medicine, Pulmonary and Critical Care, University of Virginia, 1221 Lee St, Charlottesville, Virginia 22903, USA
| | - Sarah E. Ewald
- Department of Microbiology, Immunology, and Cancer Biology, Beirne B. Carter Immunology Center, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, Virginia 22903, USA
| | - Donald Griffin
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd, Charlottesville, Virginia 22903, USA
| |
Collapse
|
40
|
Widener AE, Roberts A, Phelps EA. Granular Hydrogels for Harnessing the Immune Response. Adv Healthc Mater 2024; 13:e2303005. [PMID: 38145369 PMCID: PMC11196388 DOI: 10.1002/adhm.202303005] [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: 09/08/2023] [Revised: 11/13/2023] [Indexed: 12/26/2023]
Abstract
This review aims to understand the current progress in immune-instructive granular hydrogels and identify the key features used as immunomodulatory strategies. Published work is systematically reviewed and relevant information about granular hydrogels used throughout these studies is collected. The base polymer, microgel generation technique, polymer crosslinking chemistry, particle size and shape, annealing strategy, granular hydrogel stiffness, pore size and void space, degradability, biomolecule presentation, and drug release are cataloged for each work. Several granular hydrogel parameters used for immune modulation: porosity, architecture, bioactivity, drug release, cell delivery, and modularity, are identified. The authors found in this review that porosity is the most significant factor influencing the innate immune response to granular hydrogels, while incorporated bioactivity is more significant in influencing adaptive immune responses. Here, the authors' findings and summarized results from each section are presented and suggestions are made for future studies to better understand the benefits of using immune-instructive granular hydrogels.
Collapse
Affiliation(s)
- Adrienne E Widener
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr., Gainesville, 32611, USA
| | - Abilene Roberts
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr., Gainesville, 32611, USA
| | - Edward A Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr., Gainesville, 32611, USA
| |
Collapse
|
41
|
Gudima A, Hesselbarth D, Li G, Riabov V, Michel J, Liu Q, Schmuttermaier C, Jiao Z, Sticht C, Jawhar A, Obertacke U, Klüter H, Vrana NE, Kzhyshkowska J. Titanium induces proinflammatory and tissue-destructive responses in primary human macrophages. J Leukoc Biol 2024; 116:706-725. [PMID: 38512961 DOI: 10.1093/jleuko/qiae072] [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: 01/17/2023] [Revised: 02/06/2024] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Implants and medical devices are efficient and practical therapeutic solutions for a multitude of pathologies. Titanium and titanium alloys are used in orthopedics, dentistry, and cardiology. Despite very good mechanical properties and corrosion resistance, titanium implants can fail due to inflammatory or tissue degradation-related complications. Macrophages are major immune cells that control acceptance of failure of the implant. In this study, for the first time, we have performed a systematic analysis of the response of differentially activated human macrophages, M(Control), M(IFNγ), and M(IL-4), to the polished and porous titanium surfaces in order to identify the detrimental effect of titanium leading to the tissue destruction and chronic inflammation. Transcriptome analysis revealed that the highest number of differences between titanium and control settings are found in M(IL-4) that model healing type of macrophages. Real-time quantitative polymerase chain reaction analysis confirmed that both polished and porous titanium affected expression of cytokines, chitinases/chitinase-like proteins, and matrix metalloproteinases (MMPs). Titanium-induced release and activation of MMP7 by macrophages was enhanced by fibroblasts in both juxtacrine and paracrine cell interaction models. Production of titanium-induced MMPs and cytokines associated with chronic inflammation was independent of the presence of Staphylococcus aureus. MMP7, one of the most pronounced tissue-destroying factors, and chitinase-like protein YKL-40 were expressed in CD68+ macrophages in peri-implant tissues of patients with orthopedic implants. In summary, we demonstrated that titanium induces proinflammatory and tissue-destructing responses mainly in healing macrophages, and the detrimental effects of titanium surfaces on implant-adjacent macrophages are independent on the bacterial contamination.
Collapse
Affiliation(s)
- Alexandru Gudima
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - David Hesselbarth
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Clinic for Cardiology and Angiology, University Heart Centre Freiburg-Bad Krozingen, Freiburg, Germany
| | - Guanhao Li
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Vladimir Riabov
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
| | - Julia Michel
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Quan Liu
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christina Schmuttermaier
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Zhen Jiao
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Carsten Sticht
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany
| | - Ahmed Jawhar
- Department of Orthopaedics and Trauma Surgery, University Medical Center Mannheim, University of Heidelberg, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany
| | - Udo Obertacke
- Department of Orthopaedics and Trauma Surgery, University Medical Center Mannheim, University of Heidelberg, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany
| | - Harald Klüter
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Nihal Engin Vrana
- SPARTHA Medical, Strasbourg, France
- Department of Biomaterials and Bioengineering, UMR_S1121, Biomaterials and Bioengineering, INSERM and University of Strasburg, Strasbourg, France
| | - Julia Kzhyshkowska
- Institute for Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| |
Collapse
|
42
|
Goel A, Tathireddy H, Wang SH, Vu HH, Puy C, Hinds MT, Zonies D, McCarty OJ, Shatzel JJ. Targeting the Contact Pathway of Coagulation for the Prevention and Management of Medical Device-Associated Thrombosis. Semin Thromb Hemost 2024; 50:989-997. [PMID: 37044117 PMCID: PMC11069398 DOI: 10.1055/s-0043-57011] [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] [Indexed: 04/14/2023]
Abstract
Hemorrhage remains a major complication of anticoagulants, with bleeding leading to serious and even life-threatening outcomes in rare settings. Currently available anticoagulants target either multiple coagulation factors or specifically coagulation factor (F) Xa or thrombin; however, inhibiting these pathways universally impairs hemostasis. Bleeding complications are especially salient in the medically complex population who benefit from medical devices. Extracorporeal devices-such as extracorporeal membrane oxygenation, hemodialysis, and cardiac bypass-require anticoagulation for optimal use. Nonetheless, bleeding complications are common, and with certain devices, highly morbid. Likewise, pharmacologic prophylaxis to prevent thrombosis is not commonly used with many medical devices like central venous catheters due to high rates of bleeding. The contact pathway members FXI, FXII, and prekallikrein serve as a nexus, connecting biomaterial surface-mediated thrombin generation and inflammation, and may represent safe, druggable targets to improve medical device hemocompatibility and thrombogenicity. Recent in vivo and clinical data suggest that selectively targeting the contact pathway of coagulation through the inhibition of FXI and FXII can reduce the incidence of medical device-associated thrombotic events, and potentially systemic inflammation, without impairing hemostasis. In the following review, we will outline the current in vivo and clinical data encompassing the mechanism of action of drugs targeting the contact pathway. This new class of inhibitors has the potential to herald a new era of effective and low-risk anticoagulation for the management of patients requiring the use of medical devices.
Collapse
Affiliation(s)
- Abhishek Goel
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Harsha Tathireddy
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Si-Han Wang
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Helen H. Vu
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Cristina Puy
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Monica T. Hinds
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - David Zonies
- Department of Surgery, Oregon Health and Science University, Portland, Oregon
| | - Owen J.T. McCarty
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Joseph J. Shatzel
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
43
|
Timmerman R, Allison S, Dolivo D, Jagasia P, Gargiulo K, Battu S, Hong SJ, Helenowski I, Kim JY, Fracol M. Cosmetic implant placement in the female breast yields an altered local and systemic immune response: evidence for breast cancer immunosurveillance. Plast Reconstr Surg 2024; 155:00006534-990000000-02454. [PMID: 39344008 DOI: 10.1097/prs.0000000000011780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
BACKGROUND Women with cosmetic implants have lower rates of future breast cancer than the general population. We hypothesized the implant foreign body response could induce a local protective anti-cancer immunosurveillance. We expanded on our previous finding which showed women with breast implants have elevated antibody responses to certain breast cancer proteins. METHODS Blood samples and breast tissue were collected from women undergoing first time breast augmentation (implant-naive, IN) and revision breast augmentation (implant-exposed, IE). Sera were collected and antibody levels to common breast cancer proteins were quantified by enzyme-linked immunosorbent assay (ELISA). RT-PCR was performed on breast tissue samples to quantify immune-related gene expression levels between IN and IE. Bulk RNA sequencing was performed to identify differentially expressed genes and altered signaling pathways in the breasts of IN versus IE. RESULTS In total, 188 patients were recruited (117 IN, 71 IE). Data demonstrated that IE patients had higher levels of antibodies to MUC-1, ER, and mammaglobin A compared to IN patients. MUC-1 expression was found to be higher in IE compared to IN breast tissue. RNA-seq analysis demonstrated upregulated pathways in IE breast tissue for B cell activation and development, Th2 related genes, T cell activation, chemotactic factors, and responses to estrogen. CONCLUSION This is the first study to demonstrate that peri-implant inflammation extends beyond the implant capsule to breast parenchyma. Women with breast implants have more activated B cells in the breast parenchyma and elevated antibody responses to breast cancer antigen.
Collapse
Affiliation(s)
- Ramsey Timmerman
- Division of Plastic Surgery, Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | - Sophia Allison
- Division of Plastic Surgery, Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | - David Dolivo
- Division of Plastic Surgery, Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | - Puja Jagasia
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kristine Gargiulo
- Division of Plastic Surgery, Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | - Shreya Battu
- Division of Plastic Surgery, Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | - Seok Jong Hong
- Division of Plastic Surgery, Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | - Irene Helenowski
- Division of Plastic Surgery, Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | - John Ys Kim
- Division of Plastic Surgery, Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| | - Megan Fracol
- Division of Plastic Surgery, Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
44
|
Silva CF, Felzemburgh VDA, Moreno AD, Meneses JVL, Barbosa ADA, Barreto IC, Miguel FB. Experimental protocol for evaluation of biomaterials in an in-vivo silicone implant coverage. Acta Cir Bras 2024; 39:e396724. [PMID: 39356935 PMCID: PMC11441155 DOI: 10.1590/acb396724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/15/2024] [Indexed: 10/04/2024] Open
Abstract
PURPOSE To describe an experimental surgical model in rats using a dual-plane technique for evaluation of biomaterials in an in-vivo silicone implant coverage. METHODS This study was developed following the ISO 10993-6 standard. In this study, 40 male Wistar rats weighing between 250 and 350 g were used, distributed into two groups: experimental, biomaterial superimposed on the minimammary prosthesis (MP); and control, MP without implantation of the biomaterial, with eight animals at each biological point: 1, 2, 4, 12, and 26 weeks. Thus, at the end of biological points (1, 2, 4, 12, and 26 weeks; n = 8 animals per week), the tissue specimens achieved were fixed in buffered formalin and stained with hematoxylin-eosin. RESULTS Macroscopically, throughout the study, no postoperative complications were apparent. In the histological analysis, it was possible to observe the evolution of the inflammatory response, tissue repair, and fibrous capsule during the biological points. CONCLUSIONS The experimental model described in this study proved to be suitable for evaluating the biomaterial used in the coverage of breast silicone implants.
Collapse
Affiliation(s)
- Chenia Frutuoso Silva
- Universidade Federal da Bahia – Institute of Health Sciences – Laboratory of Tissue Bioengineering and Biomaterials – Salvador (BA) – Brazil
- Universidade Federal da Bahia – Post-graduation Program in Interactive Processes of the Organs and Systems – Salvador (BA) – Brazil
| | - Victor de Araújo Felzemburgh
- Universidade Federal da Bahia – Institute of Health Sciences – Laboratory of Tissue Bioengineering and Biomaterials – Salvador (BA) – Brazil
| | - Amanda Dourado Moreno
- Universidade Federal da Bahia – Institute of Health Sciences – Laboratory of Tissue Bioengineering and Biomaterials – Salvador (BA) – Brazil
| | | | | | - Isabela Cerqueira Barreto
- Universidade Federal da Bahia – Institute of Health Sciences – Laboratory of Tissue Bioengineering and Biomaterials – Salvador (BA) – Brazil
- Universidade Federal da Bahia – Post-graduation Program in Interactive Processes of the Organs and Systems – Salvador (BA) – Brazil
| | - Fúlvio Borges Miguel
- Universidade Federal da Bahia – Institute of Health Sciences – Laboratory of Tissue Bioengineering and Biomaterials – Salvador (BA) – Brazil
- Universidade Federal da Bahia – Post-graduation Program in Interactive Processes of the Organs and Systems – Salvador (BA) – Brazil
| |
Collapse
|
45
|
Xue JD, Gao J, Tang AF, Feng C. Shaping the immune landscape: Multidimensional environmental stimuli refine macrophage polarization and foster revolutionary approaches in tissue regeneration. Heliyon 2024; 10:e37192. [PMID: 39296009 PMCID: PMC11408064 DOI: 10.1016/j.heliyon.2024.e37192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024] Open
Abstract
In immunology, the role of macrophages extends far beyond their traditional classification as mere phagocytes; they emerge as pivotal architects of the immune response, with their function being significantly influenced by multidimensional environmental stimuli. This review investigates the nuanced mechanisms by which diverse external signals ranging from chemical cues to physical stress orchestrate macrophage polarization, a process that is crucial for the modulation of immune responses. By transitioning between pro-inflammatory (M1) and anti-inflammatory (M2) states, macrophages exhibit remarkable plasticity, enabling them to adapt to and influence their surroundings effectively. The exploration of macrophage polarization provides a compelling narrative on how these cells can be manipulated to foster an immune environment conducive to tissue repair and regeneration. Highlighting cutting-edge research, this review presents innovative strategies that leverage the dynamic interplay between macrophages and their environment, proposing novel therapeutic avenues that harness the potential of macrophages in regenerative medicine. Moreover, this review critically evaluates the current challenges and future prospects of translating macrophage-centered strategies from the laboratory to clinical applications.
Collapse
Affiliation(s)
- Jing-Dong Xue
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jing Gao
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ai-Fang Tang
- Department of Geratology, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Chao Feng
- Department of Reproductive Medicine, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200030, China
| |
Collapse
|
46
|
Chang L, Luo Y, Li W, Liu F, Guo J, Dai B, Tong W, Qin L, Wang J, Xu J. A comparative study on the effects of biodegradable high-purity magnesium screw and polymer screw for fixation in epiphyseal trabecular bone. Regen Biomater 2024; 11:rbae095. [PMID: 39346687 PMCID: PMC11427752 DOI: 10.1093/rb/rbae095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/11/2024] [Accepted: 07/08/2024] [Indexed: 10/01/2024] Open
Abstract
With mechanical strength close to cortical bone, biodegradable and osteopromotive properties, magnesium (Mg)-based implants are promising biomaterials for orthopedic applications. However, during the degradation of such implants, there are still concerns on the potential adverse effects such as formation of cavities, osteolytic phenomena and chronic inflammation. Therefore, to transform Mg-based implants into clinical practice, the present study evaluated the local effects of high-purity Mg screws (HP-Mg, 99.99 wt%) by comparing with clinically approved polylactic acid (PLA) screws in epiphyseal trabecular bone of rabbits. After implantation of screws at the rabbit distal femur, bone microstructural, histomorphometric and biomechanical properties were measured at various time points (weeks 4, 8 and 16) using micro-CT, histology and histomorphometry, micro-indentation and scanning electron microscope. HP-Mg screws promoted peri-implant bone ingrowth with higher bone mass (BV/TV at week 4: 0.189 ± 0.022 in PLA group versus 0.313 ± 0.053 in Mg group), higher biomechanical properties (hardness at week 4: 35.045 ± 1.000 HV in PLA group versus 51.975 ± 2.565 HV in Mg group), more mature osteocyte LCN architecture, accelerated bone remodeling process and alleviated immunoreactive score (IRS of Ram11 at week 4: 5.8 ± 0.712 in PLA group versus 3.75 ± 0.866 in Mg group) as compared to PLA screws. Furthermore, we conducted finite element analysis to validate the superiority of HP-Mg screws as orthopedic implants by demonstrating reduced stress concentration and uniform stress distribution around the bone tunnel, which led to lower risks of trabecular microfractures. In conclusion, HP-Mg screws demonstrated greater osteogenic bioactivity and limited inflammatory response compared to PLA screws in the epiphyseal trabecular bone of rabbits. Our findings have paved a promising way for the clinical application of Mg-based implants.
Collapse
Affiliation(s)
- Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Ying Luo
- School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou, Guangdong, 510000, China
| | - Weirong Li
- Dongguan Eontec Co., Ltd, Dongguan, Guangdong, 510730, China
| | - Fangfei Liu
- Dongguan Eontec Co., Ltd, Dongguan, Guangdong, 510730, China
| | - Jiaxin Guo
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Jiali Wang
- School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou, Guangdong, 510000, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| |
Collapse
|
47
|
Wang M, Hong Y, Fu X, Sun X. Advances and applications of biomimetic biomaterials for endogenous skin regeneration. Bioact Mater 2024; 39:492-520. [PMID: 38883311 PMCID: PMC11179177 DOI: 10.1016/j.bioactmat.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 06/18/2024] Open
Abstract
Endogenous regeneration is becoming an increasingly important strategy for wound healing as it facilitates skin's own regenerative potential for self-healing, thereby avoiding the risks of immune rejection and exogenous infection. However, currently applied biomaterials for inducing endogenous skin regeneration are simplistic in their structure and function, lacking the ability to accurately mimic the intricate tissue structure and regulate the disordered microenvironment. Novel biomimetic biomaterials with precise structure, chemical composition, and biophysical properties offer a promising avenue for achieving perfect endogenous skin regeneration. Here, we outline the recent advances in biomimetic materials induced endogenous skin regeneration from the aspects of structural and functional mimicry, physiological process regulation, and biophysical property design. Furthermore, novel techniques including in situ reprograming, flexible electronic skin, artificial intelligence, single-cell sequencing, and spatial transcriptomics, which have potential to contribute to the development of biomimetic biomaterials are highlighted. Finally, the prospects and challenges of further research and application of biomimetic biomaterials are discussed. This review provides reference to address the clinical problems of rapid and high-quality skin regeneration.
Collapse
Affiliation(s)
- Mengyang Wang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
| | - Yiyue Hong
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100089, PR China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| |
Collapse
|
48
|
Díez-Tercero L, Bosch-Rué È, Bosch BM, Rojas-Márquez R, Caballé-Serrano J, Delgado LM, Pérez RA. Engineering a microparticle-loaded rough membrane for guided bone regeneration modulating osteoblast response without inducing inflammation. Colloids Surf B Biointerfaces 2024; 241:113994. [PMID: 38850744 DOI: 10.1016/j.colsurfb.2024.113994] [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: 03/12/2024] [Revised: 05/14/2024] [Accepted: 05/26/2024] [Indexed: 06/10/2024]
Abstract
Guided bone regeneration (GBR) is a widely used procedure that prevents the fast in-growth of soft tissues into bone defect. Among the different types of membranes, the use of collagen membranes is the gold standard. However, these membranes are implanted in tissue location where a severe acute inflammation will occur and can be negatively affected. The aim of this study was to develop a collagen-based membrane for GBR that incorporated alginate-hydroxyapatite microparticles. Membranes were manufactured using collagen type I and gelatin and alginate-hydroxyapatite microparticles. Membranes were assessed in terms of topography by scanning electron microscopy and confocal microscopy; stability by swelling after an overnight incubation in saline and enzymatic degradation against collagenase and mechanical properties by tensile tests. Furthermore, the biological response was assessed with SaOs-2 cells and THP-1 macrophages to determine alkaline phosphatase activity and inflammatory cytokine release. Our results showed that the incorporation of different percentages of these microparticles could induce changes in the surface topography. When the biological response was analyzed, either membranes were not cytotoxic to THP-1 macrophages or to SaOs-2 cells and they did not induce the release of pro-inflammatory cytokines. However, the different surface topographies did not induce changes in the macrophage morphology and the release of pro- and anti-inflammatory cytokines, suggesting that the effect of surface roughness on macrophage behavior could be dependent on other factors such as substrate stiffness and composition. Collagen-gelatin membranes with embedded alginate-hydroxyapatite microparticles increased ALP activity, suggesting a positive effect of them on bone regeneration, remaining unaffected the release of pro- and anti-inflammatory cytokines.
Collapse
Affiliation(s)
- Leire Díez-Tercero
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Èlia Bosch-Rué
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Begoña M Bosch
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Raquel Rojas-Márquez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Jordi Caballé-Serrano
- Department of Oral and Maxillofacial Surgery, Universitat Internacional de Catalunya, Barcelona, Spain; Department of Periodontology, School of Dental Medicine - University of Bern, Bern, Switzerland
| | - Luis M Delgado
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain.
| | - Román A Pérez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain; Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona, Spain.
| |
Collapse
|
49
|
Niebles Navas AF, Araujo-Rodríguez DG, Valencia-Llano CH, Insuasty D, Delgado-Ospina J, Navia-Porras DP, Zapata PA, Albis A, Grande-Tovar CD. Lyophilized Polyvinyl Alcohol and Chitosan Scaffolds Pre-Loaded with Silicon Dioxide Nanoparticles for Tissue Regeneration. Molecules 2024; 29:3850. [PMID: 39202929 PMCID: PMC11356782 DOI: 10.3390/molecules29163850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/31/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Materials with a soft tissue regenerative capacity can be produced using biopolymer scaffolds and nanomaterials, which allow injured tissue to recover without any side effects or limitations. Four formulations were prepared using polyvinyl alcohol (PVA) and chitosan (CS), with silicon dioxide nanoparticles (NPs-SiO2) incorporated using the freeze-drying method at a temperature of -50 °C. TGA and DSC showed no change in thermal degradation, with glass transition temperatures around 74 °C and 77 °C. The interactions between the hydroxyl groups of PVA and CS remained stable. Scanning electron microscopy (SEM) indicated that the incorporation of NPs-SiO2 complemented the freeze-drying process, enabling the dispersion of the components on the polymeric matrix and obtaining structures with a small pore size (between 30 and 60 μm) and large pores (between 100 and 160 μm). The antimicrobial capacity analysis of Gram-positive and Gram-negative bacteria revealed that the scaffolds inhibited around 99% of K. pneumoniae, E. cloacae, and S. aureus ATCC 55804. The subdermal implantation analysis demonstrated tissue growth and proliferation, with good biocompatibility, promoting the healing process for tissue restoration through the simultaneous degradation and formation of type I collagen fibers. All the results presented expand the boundaries in tissue engineering and regenerative medicine by highlighting the crucial role of nanoparticles in optimizing scaffold properties.
Collapse
Affiliation(s)
- Andrés Felipe Niebles Navas
- Grupo de Investigación de Fotoquímica y Fotobiología, Universidad del Atlántico, Carrera 30 Número 8-49, Puerto Colombia 081008, Colombia
| | - Daniela G Araujo-Rodríguez
- Grupo de Investigación de Fotoquímica y Fotobiología, Universidad del Atlántico, Carrera 30 Número 8-49, Puerto Colombia 081008, Colombia
| | - Carlos-Humberto Valencia-Llano
- Grupo Biomateriales Dentales, Escuela de Odontología, Universidad del Valle, Calle 4B Número 36-00, Cali 760001, Colombia
| | - Daniel Insuasty
- Departamento de Química y Biología, División de Ciencias Básicas, Universidad del Norte, Km 5 Vía Puerto Colombia, Barranquilla 081007, Colombia
| | - Johannes Delgado-Ospina
- Grupo de Investigación Biotecnología, Facultad de Ingeniería, Universidad de San Buenaventura Cali, Carrera 122 Número 6-65, Cali 760001, Colombia
| | - Diana Paola Navia-Porras
- Grupo de Investigación Biotecnología, Facultad de Ingeniería, Universidad de San Buenaventura Cali, Carrera 122 Número 6-65, Cali 760001, Colombia
| | - Paula A Zapata
- Grupo de Polímeros, Facultad de Química y Biología, Universidad de Santiago de Chile (USACH), Santiago 9170020, Chile
| | - Alberto Albis
- Grupo de Investigación en Bioprocesos, Facultad de Ingeniería, Universidad del Atlántico, Carrera 30 Número 8-49, Puerto Colombia 081008, Colombia
| | - Carlos David Grande-Tovar
- Grupo de Investigación de Fotoquímica y Fotobiología, Universidad del Atlántico, Carrera 30 Número 8-49, Puerto Colombia 081008, Colombia
| |
Collapse
|
50
|
Deng H, Guan Y, Dong Q, An R, Wang J. Chitosan-based biomaterials promote bone regeneration by regulating macrophage fate. J Mater Chem B 2024; 12:7480-7496. [PMID: 39016095 DOI: 10.1039/d3tb02563b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
The development of various osteogenic biomaterials has not only promoted the development of bone tissue engineering but also provided more possibilities for bone defect repair. However, most previous studies have focused on the interaction of biomaterials on endogenous or exogenous stem cells involved in the bone regeneration process while neglecting the effect of changes in the immune microenvironment of bone defect sites on bone regeneration after biomaterial implantation into the host. With the development of bone immunology, the role of various immune cells, especially macrophages, in bone regeneration has gradually attracted the attention of researchers. An increasing number of studies have begun to target macrophages to better promote bone regeneration by modulating the fate of macrophages in a spatiotemporally ordered manner to mimic the changes in the immune microenvironment of bone defect sites during the natural repair process of bone tissue. Chitosan is one of the most abundant natural polysaccharides in the world. In recent years, various chitosan-based biomaterials have been widely used in macrophage fate modulation and bone regeneration. In this review, we review the interaction between macrophages and scaffold materials, general information about chitosan, the modulation of macrophage fate by chitosan-based biomaterials, and their application in bone regeneration.
Collapse
Affiliation(s)
- Huiling Deng
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China.
| | - Yuanyuan Guan
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China.
| | - Quping Dong
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China.
| | - Ran An
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China.
| | - Jiecong Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China.
| |
Collapse
|