1
|
Li S, Man Z, Zuo K, Zhang L, Zhang T, Xiao G, Lu Y, Li W, Li N. Advancement in smart bone implants: the latest multifunctional strategies and synergistic mechanisms for tissue repair and regeneration. Bioact Mater 2025; 51:333-382. [PMID: 40491688 PMCID: PMC12146007 DOI: 10.1016/j.bioactmat.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/10/2025] [Accepted: 05/07/2025] [Indexed: 06/11/2025] Open
Abstract
Artificial implants have consistently been recognized as the most effective clinical strategy for repairing bone fractures and defects, particularly in orthopedics and stomatology. Nowadays, the focus of bone repair has shifted from basic fixation and structural restoration to the reconstruction of multifunctional "live" tissue to mimic the natural bone microenvironment. However, developing the smart implants with ideal osteogenesis-related multi-functions remains challenging, as the effects of physicochemical properties of implant materials on intracellular signaling, stem cell niches, and tissue regeneration are not yet fully understood. Herein, we systematically explore recent advancements in innovative strategies for bone repair and regeneration, revealing the significance of the smart implants that closely mimic the natural structure and function of bone tissue. Adaptation to patient-oriented osteogenic microenvironments, dynamic osteoblastogenesis-osteoclastogenesis balance, antibacterial/bactericidal capacity, vascularization, and osteoimmunomodulatory capacity and their regulatory mechanisms achieved by biomaterials design and functional modifications are thoroughly summarized and analyzed. Notably, the popular research on multifunctional platforms with synergetic interactions between different functions and treatment of complex clinical issues, including the emerging neurogenic bone repair, is also significantly discussed for developing more intelligent implants. By summarizing recent research efforts, this review proposes the latest multifunctional strategies and synergistic mechanisms of smart bone implants, aiming to provide better bone defect repair applications that more closely mimic the natural bone tissue.
Collapse
Affiliation(s)
- Shishuo Li
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong Province, 250021, PR China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, 250117, PR China
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong Province, 250021, PR China
- Graduate School of Dalian Medical University, Dalian, Liaoning Province, 116044, PR China
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, Jiangsu Province, 213003, PR China
| | - Zhentao Man
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong Province, 250021, PR China
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, 250021, PR China
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong Institute of Endocrine and Metabolic Diseases, Ji'nan, Shandong, 250062, PR China
| | - Kangqing Zuo
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong Province, 250021, PR China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, 250117, PR China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, 250117, PR China
| | - Linbo Zhang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong Province, 250021, PR China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, 250117, PR China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, 250117, PR China
| | - Taixing Zhang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong Province, 250021, PR China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, 250117, PR China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, 250117, PR China
| | - Guiyong Xiao
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Ji'nan, 250061, PR China
- School of Materials Science and Engineering, Shandong University, Ji'nan, 250061, PR China
| | - Yupeng Lu
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Ji'nan, 250061, PR China
- School of Materials Science and Engineering, Shandong University, Ji'nan, 250061, PR China
| | - Wei Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong Province, 250021, PR China
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, 250021, PR China
| | - Ningbo Li
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji'nan, Shandong Province, 250021, PR China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, 250117, PR China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong Province, 250117, PR China
| |
Collapse
|
2
|
Therriault MA, Kottapalli S, Artsen A, Knight K, King G, Meyn L, Brown BN, Moalli PA. Profiling of the macrophage response to polypropylene mesh burden in vivo. Biomaterials 2025; 318:123177. [PMID: 39961254 DOI: 10.1016/j.biomaterials.2025.123177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/01/2024] [Accepted: 02/07/2025] [Indexed: 03/05/2025]
Abstract
Pelvic organ prolapse (POP) surgical repair with polypropylene mesh (PPM) offers improved anatomical outcomes compared to reconstruction using native tissue. However, PPM repair is hampered by complications, most commonly pain or mesh exposure, occurring in over 10 % of cases. This maladaptive response is, in part, attributed to the host response to a foreign material. Previous studies have demonstrated that mesh properties, such as weight, pore size, and porosity, influence downstream outcomes. In addition, computational models and in vivo mechanistic studies demonstrate that mesh deforms after tensioning in prolapse surgery resulting in collapsed pores and wrinkles. To further investigate the role of pore collapse in mesh complications, PPM was implanted flat, or in configurations that would deform upon tensioning in a POP repair surgery using a non-human primate model. After twelve weeks, we analyzed mesh-tissue complexes to characterize the overall host response, profile the macrophage response, and observe the influence of macrophages in downstream healing outcomes that may lead to complications. The results confirm that mesh deformations reproduce mesh exposure and thinning of vagina. In the PPM configurations with the greatest deformation, mesh burden was the highest, which resulted in an overall decrease in the number of cells within the implantation site. Among the cells that were present, we observed a predominance of M1 pro-inflammatory macrophages. While flat mesh was associated with an organized cellular response, deformed mesh led to an increasingly disorganized response as mesh burden increased. Nearly half of the responding macrophages expressed markers associated both with M1 and M2 phenotypes concurrently, suggesting the possibility of newly recruited macrophages responding even 12 weeks after implantation and/or a repetitive microinjury in which macrophages are continuously recruited and polarized without resolution of the host response. Biochemically, we observed a predominantly M1 pro-inflammatory signaling environment and decreased collagen content as a response to implanted mesh. This study evidences the importance of PPM mesh properties, which may alter mesh burden upon tensioning and impact downstream healing outcomes and emphasizes the need for devices that maintain their geometry following implantation in POP surgical repair.
Collapse
Affiliation(s)
- Marrisa A Therriault
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Magee Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Srividya Kottapalli
- Magee Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amanda Artsen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA; Magee Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA; Division of Urogynecology & Reconstructive Pelvic Surgery, University of Pittsburgh Medical Center Magee-Womens Hospital, Pittsburgh, PA, USA
| | - Katrina Knight
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA; Magee Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gabrielle King
- Magee Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leslie Meyn
- Magee Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bryan N Brown
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA; Magee Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Pamela A Moalli
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA; Magee Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA; Division of Urogynecology & Reconstructive Pelvic Surgery, University of Pittsburgh Medical Center Magee-Womens Hospital, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Kibet M, Abebayehu D. Crosstalk between T cells and fibroblasts in biomaterial-mediated fibrosis. Matrix Biol Plus 2025; 26:100172. [PMID: 40226302 PMCID: PMC11986236 DOI: 10.1016/j.mbplus.2025.100172] [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: 09/24/2024] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
Biomaterial implants are a critical aspect of our medical therapies and biomedical research and come in various forms: stents, implantable glucose sensors, orthopedic implants, silicone implants, drug delivery systems, and tissue engineered scaffolds. Their implantation triggers a series of biological responses that often times lead to the foreign body response and subsequent fibrotic encapsulation, a dense ECM-rich capsule that isolates the biomaterial and renders it ineffective. These responses lead to the failure of biomaterials and is a major hurdle to overcome and in promoting their success. Much attention has been given to macrophage populations for the inflammatory component of these responses to biomaterials but recent work has identified an important role of T cells and their ability to modulate fibroblast activity and vice versa. In this review, we focus on T cell-fibroblast crosstalk by exploring T cell subsets, critical signaling pathways, and fibroblast populations that have been shown to dictate biomaterial-mediated fibrosis. We then highlight emerging technologies and model systems that enable new insights and avenues to T cell-fibroblast crosstalk that will improve biomaterial outcomes.
Collapse
Affiliation(s)
- Mathew Kibet
- Department of Biomedical Engineering, School of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, United States
| | - Daniel Abebayehu
- Department of Biomedical Engineering, School of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, United States
| |
Collapse
|
4
|
Sun J, Xie W, Wu Y, Li Z, Li Y. Accelerated Bone Healing via Electrical Stimulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404190. [PMID: 39115981 DOI: 10.1002/advs.202404190] [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: 04/19/2024] [Revised: 07/01/2024] [Indexed: 08/10/2024]
Abstract
Piezoelectric effect produces an electrical signal when stress is applied to the bone. When the integrity of the bone is destroyed, the biopotential within the defect site is reduced and several physiological responses are initiated to facilitate healing. During the healing of the bone defect, the bioelectric potential returns to normal levels. Treatment of fractures that exceed innate regenerative capacity or exhibit delayed healing requires surgical intervention for bone reconstruction. For bone defects that cannot heal on their own, exogenous electric fields are used to assist in treatment. This paper reviews the effects of exogenous electrical stimulation on bone healing, including osteogenesis, angiogenesis, reduction in inflammation and effects on the peripheral nervous system. This paper also reviews novel electrical stimulation methods, such as small power supplies and nanogenerators, that have emerged in recent years. Finally, the challenges and future trends of using electrical stimulation therapy for accelerating bone healing are discussed.
Collapse
Affiliation(s)
- Jianfeng Sun
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yuxiang Wu
- School of Kinesiology, Jianghan University, Wuhan, Hubei, 430056, China
| | - Zhou Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| |
Collapse
|
5
|
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
|
6
|
Guan Y, Zhang M, Song J, Negrete M, Adcock T, Kandel R, Racioppi L, Gerecht S. CaMKK2 Regulates Macrophage Polarization Induced by Matrix Stiffness: Implications for Shaping the Immune Response in Stiffened Tissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417778. [PMID: 40036145 PMCID: PMC12021110 DOI: 10.1002/advs.202417778] [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: 12/30/2024] [Revised: 01/13/2025] [Indexed: 03/06/2025]
Abstract
Macrophages are essential for immune responses and maintaining tissue homeostasis, exhibiting a wide range of phenotypes depending on their microenvironment. The extracellular matrix (ECM) is a vital component that provides structural support and organization to tissues, with matrix stiffness acting as a key regulator of macrophage behavior. Using physiologically relevant 3D stiffening hydrogel models, it is found that increased matrix stiffness alone promoted macrophage polarization toward a pro-regenerative phenotype, mimicking the effect of interleukin-4(IL-4) in softer matrices. Blocking Calcium/calmodulin-dependent kinase kinase 2 (CaMKK2) selectively inhibited stiffness-induced macrophage polarization without affecting IL-4-driven pro-regenerative pathways. In functional studies, CaMKK2 deletion prevented M2-like/pro-tumoral polarization caused by matrix stiffening, which in turn hindered tumor growth. In a murine wound healing model, loss of CaMKK2 impaired matrix stiffness-mediated macrophage accumulation, ultimately disrupting vascularization. These findings highlight the critical role of CaMKK2 in the macrophage mechanosensitive fate determination and gene expression program, positioning this kinase as a promising therapeutic target to selectively modulate macrophage responses in pathologically stiff tissues.
Collapse
Affiliation(s)
- Ya Guan
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Min Zhang
- Division of Hematological Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27708USA
| | - Jiyeon Song
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Marcos Negrete
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Tyler Adcock
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Reeva Kandel
- Division of Hematological Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27708USA
| | - Luigi Racioppi
- Division of Hematological Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27708USA
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples Federico IINaplesItaly
| | - Sharon Gerecht
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
7
|
Du F, Hou M, Lu S, Ding X, Zhang L, Du Y, An Z, Cai W, Zhao L, Wu W, Cao Z. Toxicity enhancement of microplastics released from food containers through thermal aging: Absorbing more serum proteins thus activating the innate immune response via actin polymerization. ENVIRONMENT INTERNATIONAL 2025; 197:109358. [PMID: 40049044 DOI: 10.1016/j.envint.2025.109358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/01/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
This study examined the effects of hot high-fat simulants on the physicochemical properties of microplastics (MPs) from polypropylene (PP)-, low-density polyethylene (LDPE)-, and polylactic acid (PLA)-based single-use food container (SUFC) leachates and those of aging on their immunomodulatory effectors. Scenario studies have demonstrated that MPs were released from these three types of SUFCs. LDPE- and PLA-based SUFCs also released cellulose. Among the SUFCs, only the PP leachates particles exhibited a new absorption peak at 1725 cm-1, which aging phenomenon may be attributed to the presence of unstable tertiary carbon atoms. Subsequently, we investigated the immunomodulatory effects of removing additive both PP and thermal-aged PP with polystyrene (PS) and carboxyl-modified PS (PS-COOH) polymer backbones as reference materials. The findings indicated that thermal-aged PP and PS-COOH induced comparable innate immune responses, with PS-COOH particles exhibiting a similar size to SUFC percolates. Consequently, PS and PS-COOH were selected as original and thermal-aged MPs, respectively, to evaluate the effects of aging on innate immunity. The results revealed thata protein corona formed on both particle types, with more protein adsorption observed on PS-COOH particles. The complex enhanced the phagocytosis of RAW264.7 macrophages and increased the expression of pro-inflammatory genes NOS2 and TNF-α through an actin polymerization cross-linking mechanism. In this study, we investigated how thermal-aged MPs affect innate immune responses using PS-COOH as a model system, emphasizing the importance of a comprehensive safety evaluations of MPs.
Collapse
Affiliation(s)
- Fang Du
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Normal University, Xinxiang 453007, China
| | - Meiqian Hou
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Normal University, Xinxiang 453007, China
| | - Song Lu
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Normal University, Xinxiang 453007, China
| | - Xiaotian Ding
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Normal University, Xinxiang 453007, China
| | - Ling Zhang
- School of Public Health, Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, Xinxiang Medical University, Xinxiang 453003, China
| | - Yajie Du
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Normal University, Xinxiang 453007, China
| | - Zhen An
- School of Public Health, Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, Xinxiang Medical University, Xinxiang 453003, China
| | - Wenwen Cai
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Normal University, Xinxiang 453007, China; Huanghuai Laboratory, Zhengzhou, Henan 450003, China
| | - Leicheng Zhao
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Normal University, Xinxiang 453007, China; Huanghuai Laboratory, Zhengzhou, Henan 450003, China
| | - Weidong Wu
- School of Public Health, Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, Xinxiang Medical University, Xinxiang 453003, China.
| | - Zhiguo Cao
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Normal University, Xinxiang 453007, China; Huanghuai Laboratory, Zhengzhou, Henan 450003, China.
| |
Collapse
|
8
|
Yu F, Zhao X, Zhang S, Lu W, Li P, Yang W, Zhao Z. Regulation of T Cell Glycosylation by MXene/β-TCP Nanocomposite for Enhanced Mandibular Bone Regeneration. Adv Healthc Mater 2025; 14:e2404015. [PMID: 39764719 DOI: 10.1002/adhm.202404015] [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: 10/16/2024] [Revised: 12/16/2024] [Indexed: 03/04/2025]
Abstract
Immune-mediated bone regeneration driven by bone biomaterials offers a therapeutic strategy for repairing bone defects. Among 2D nanomaterials, Ti3C2Tx MXenes have garnered substantial attention for their potential in tissue regeneration. This investigation concentrates on the role of MXene nanocomposites in modulating the immune microenvironment within bone defects to facilitate bone tissue restoration. Ti3C2Tx MXenes are synthetized, incorporated into beta-tricalcium phosphate ceramics (β-TCP) nanocomposites (T-MXene), and their osteoinductive and immunomodulatory effects are evaluated. The effects of T-MXene-treated T-cells on bone marrow stromal cells (BMSCs) are explored. In addition, its therapeutic potential for bone regeneration is assessed in vivo using a critical-sized mandibular bone defect model. The underlying mechanisms by which T-MXene regulates T-cell differentiation and bone regeneration are explored via whole-transcriptome RNA sequencing. The scaffolds activate N-glycosylation in T cells, which possess anti-inflammatory and antioxidant effects, thereby inducing a pro-regenerative response. T-MXene increased the proportion of IL-4+ T cells among primary T cells and mandibular lymph nodes, ultimately promoting osteogenesis in BMSCs and injured mandibles. The distinctive function of MXene-based nanocomposites in osteoimmunomodulation provides a solid foundation for further exploration and application of MXenes as immune response modulators, potentially advancing their use in regenerative medicine.
Collapse
Affiliation(s)
- Fei Yu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xing Zhao
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Shuting Zhang
- College of Polymer Science and Engineering, Med-X Center for Materials, Sichuan University, Chengdu, 610065, China
| | - Wenxin Lu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Peilin Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Wei Yang
- College of Polymer Science and Engineering, Med-X Center for Materials, Sichuan University, Chengdu, 610065, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
9
|
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
|
10
|
do Carmo DJAC, Lazari MGT, dos Santos LCC, Costa PAC, Jesus ICG, Guatimosim S, Guimaraes PPG, Andrade SP, Campos PP. Sodium propionate decreases implant-induced foreign body response in mice. PLoS One 2025; 20:e0316764. [PMID: 39970160 PMCID: PMC11838875 DOI: 10.1371/journal.pone.0316764] [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: 08/07/2024] [Accepted: 12/16/2024] [Indexed: 02/21/2025] Open
Abstract
The short-chain fatty acid (SCFA) propionate, beyond its actions on the intestine, has been able to lower inflammation and modulate angiogenesis and fibrogenesis in pathological conditions in experimental animal models. Its effects on foreign body reaction (FBR), an abnormal healing process induced by implantation of medical devices, have not been investigated. We have evaluated the effects of sodium propionate (SP) on inflammation, neovascularization and remodeling on a murine model of implant-induced FBR. Polyether-polyurethane sponge discs implanted subcutaneously in C57BL/6 mice provided the scaffold for the formation of the fibrovascular tissue. Fifteen-day old implants of the treated group (SP, 100 mg/kg for 14 days) presented a decrease in the inflammatory response as evaluated by cellular influx (flow cytometry; Neutrophils 54%; Lymphocytes 25%, Macrophages 40%). Myeloperoxidase activity, TNF-α levels and mast cell number were also lower in the treated group relative to the control group. Angiogenesis was evaluated by blood vessel number and VEGF levels, which were downregulated by the treatment. Moreover, the number of foreign body giant cells HE (FBGC) and the thickness of the collagenous capsule were reduced by 58% and 34%, respectively. Collagen deposition inside the implant, TGF-β1 levels, α-SMA and TGF-β1 expression were also reduced. These effects may indicate that SP holds potential as a therapeutic agent for attenuating adverse remodeling processes associated with implantable devices, expanding its applications in biomedical contexts.
Collapse
Affiliation(s)
| | - Marcela Guimarães Takahashi Lazari
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Campus UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Letícia Cristine Cardoso dos Santos
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Campus UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro Augusto Carvalho Costa
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Campus UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Itamar Couto Guedes Jesus
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Campus UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Campus UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro Pires Goulart Guimaraes
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Campus UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Silvia Passos Andrade
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Campus UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Paula Peixoto Campos
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Campus UFMG, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
11
|
Pisani S, Evangelista A, Chesi L, Croce S, Avanzini MA, Dorati R, Genta I, Benazzo M, Comoli P, Conti B. Nanofibrous Scaffolds' Ability to Induce Mesenchymal Stem Cell Differentiation for Soft Tissue Regenerative Applications. Pharmaceuticals (Basel) 2025; 18:239. [PMID: 40006052 PMCID: PMC11859969 DOI: 10.3390/ph18020239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have gained recognition as a highly versatile and promising cell source for repopulating bioengineered scaffolds due to their inherent capacity to differentiate into multiple cell types. However, MSC implantation techniques have often yielded inconsistent clinical results, underscoring the need for advanced approaches to enhance their therapeutic efficacy. Recent developments in three-dimensional (3D) bioengineered scaffolds have provided a significant breakthrough by closely mimicking the in vivo environment, addressing the limitations of traditional two-dimensional (2D) cell cultures. Among these, nanofibrous scaffolds have proven particularly effective, offering an optimal 3D framework, growth-permissive substrates, and the delivery of trophic factors crucial for MSC survival and regeneration. Furthermore, the selection of appropriate biomaterials can amplify the paracrine effects of MSCs, promoting both proliferation and targeted differentiation. The synergistic combination of MSCs with nanofibrous scaffolds has demonstrated remarkable potential in achieving repair, regeneration, and tissue-specific differentiation with enhanced safety and efficacy, paving the way for routine clinical applications. In this review, we examine the most recent studies (2013-2023) that explore the combined use of MSCs and nanofibrous scaffolds for differentiation into cardiogenic, epithelial, myogenic, tendon, and vascular cell lineages. Using PubMed, we identified and analyzed 275 relevant articles based on the search terms "Nanofibers", "Electrospinning", "Mesenchymal stem cells", and "Differentiation". This review highlights the critical advancements in the use of nanofibrous scaffolds as a platform for MSC differentiation and tissue regeneration. By summarizing key findings from the last decade, it provides valuable insights for researchers and clinicians aiming to optimize scaffold design, MSC integration, and translational applications. These insights could significantly influence future research directions and the development of more effective regenerative therapies.
Collapse
Affiliation(s)
- Silvia Pisani
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Aleksandra Evangelista
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Luca Chesi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Stefania Croce
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.C.); (M.A.A.); (P.C.)
| | - Maria Antonietta Avanzini
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.C.); (M.A.A.); (P.C.)
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Marco Benazzo
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Patrizia Comoli
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.C.); (M.A.A.); (P.C.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| |
Collapse
|
12
|
Zhang L, Jiang H, Ma H. Progress in immune microenvironment, immunotherapy and prognostic biomarkers in pediatric osteosarcoma. Front Immunol 2025; 16:1548527. [PMID: 39911380 PMCID: PMC11794274 DOI: 10.3389/fimmu.2025.1548527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
Pediatric osteosarcoma, the most prevalent primary malignant bone tumor in children, is marked by aggressive progression and a generally poor prognosis. Despite advances in treatment, including multi-agent chemotherapy, survival rates remain suboptimal, with metastasis, particularly to the lungs, contributing significantly to mortality. The tumor microenvironment plays a crucial role in osteosarcoma progression, with immune cells such as tumor-associated macrophages and T lymphocytes significantly influencing tumor behavior. The immunosuppressive environment, dominated by M2 macrophages, contributes to immune evasion and poor therapeutic outcomes, though recent findings suggest the potential for reprogramming these cells to enhance immune responses. This review provides a comprehensive overview of the immune landscape in pediatric osteosarcoma, with a focus on the role of immune cells and their interactions within the tumor microenvironment (TME). It examines the impact of immune checkpoints, genetic mutations, and inflammatory pathways on osteosarcoma progression, highlighting their contribution to tumor immune evasion and disease advancement. Additionally, emerging immunotherapeutic strategies, such as immune checkpoint inhibitors, macrophage reprogramming, and antibody-based therapies, are summarized in detail, showcasing their potential to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Haoming Jiang
- Department of Orthopedics, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Haichao Ma
- Department of Pediatrics, Shenzhen University General Hospital, Shenzhen, China
| |
Collapse
|
13
|
Li M, Chen Q, Zhou M, Li X, Wang Z, Wang J. α-Ketoglutaric Acid Reprograms Macrophages by Altering Energy Metabolism to Promote the Regeneration of Small-Diameter Vascular Grafts. ACS Biomater Sci Eng 2025; 11:518-530. [PMID: 39604080 DOI: 10.1021/acsbiomaterials.4c01702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Small-diameter vascular grafts still cannot clinically replace autologous blood vessels due to high restenosis rates caused by long-term inflammatory infiltration. Foreign body reactions to vascular grafts induce macrophages to adopt the pro-inflammatory M1 phenotype, releasing inflammatory factors such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). This induces a phenotypic switch in smooth muscle cells, eventually leading to intimal hyperplasia. Herein, we constructed small-diameter artificial vascular grafts capable of modulating immune responses through the controlled release of α-ketoglutaric acid (α-KG). Our findings verify that the delivery of α-KG reprograms the macrophage phenotype from a pro-inflammatory M1 to an anti-inflammatory and pro-repair M2 phenotype by regulating the energy metabolism of the tricarboxylic acid cycle (TAC). More interestingly, the delivery of α-KG positively influences the behavior of vascular cells by enhancing the proliferation of human umbilical vein endothelial cells (HUVECs) and inhibiting the expansion of mouse aortic vascular smooth muscle cells (MOVAS), thereby reducing vascular restenosis. In vivo evaluation in rabbit carotid artery replacement confirms the optimal performance of α-KG-doped vascular grafts in terms of endothelial coverage and long-term patency. Collectively, our work presents a promising approach for creating artificial vascular grafts with inflammatory regulation to ensure rapid endothelialization and sustained patency.
Collapse
Affiliation(s)
- Mengyu Li
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qi Chen
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mengxue Zhou
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaomeng Li
- National Center for International Research of Micro-Nano Molding Technology, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Zihao Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Jianglin Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
14
|
Yin J, Liao Y, Liu S, Che B, Zhu H, Yang B, Shi B. Titanium nanotubes modulate immunophenotyping and cytokine secretion of T cells via IL-17A: a bioinformatic analysis and experimental validation. Front Immunol 2025; 15:1381158. [PMID: 39840051 PMCID: PMC11747796 DOI: 10.3389/fimmu.2024.1381158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Object We aim to explore the immunomodulatory properties of T cells on different titanium nanotubes and the key immunological factors involved in this process. Methods Transcriptome data from GEO database of healthy people and healthy implants were used to analyze cell infiltration and factor distribution of adaptive immune using bioinformatics tools. T cells from activated rat were cultured on titanium nanotubes that were prepared by anodization with different diameters (P-0, NT15-30 nm, NT40-100 nm, NT70-200 nm). The proliferation and expressions of the main transcription factors and cytokines of T-cells were detected. Magnetic bead sorting of CD3+ T cells and transcriptome sequencing were performed to explore the signaling pathways and key immune factors that may influence the related immune responses. Results Bioinformatics analysis showed that healthy peri-implant tissues were enriched by the most of T-cell subtypes. T-cell-mediated adaptive immunological responses involved IL-17A. On the third day, the NT15 and NT40 groups showed significantly higher pro-proliferative effects than the NT70 group (P<0.05). Notably, the NT40 group exhibited the lowest T-bet expression (P<0.05) along with the highest levels of Rorγt, Gata3, and Foxp3(P<0.05), followed by the NT15 group. Additionally, the NT40 group demonstrated reduced RANKL, TNF-α, and IL-6 (P<0.05) and increased OPG and IL-10 (P<0.05). Meanwhile, the NT15 group had lower IFN-γ expression(P>0.05) but higher IL-4, and TGF-β1 expressions(P<0.05). Differential expressed genes (DGEs) of T-cell related to the morphologies of titanium nanotubes were mostly enriched in the IL-17 signaling pathway mediated by IL-17A/F. Gene and protein expressions indicated that the NT40 group had the highest secretion in IL-17A of T cells. Conclusion Titanium nanotube morphologies in medium (100 nm) and small (30 nm) sizes significantly influence T cell differentiation and immune factor secretion, with T-cell-derived IL-17A likely playing a key regulatory role.
Collapse
Affiliation(s)
- Jingju Yin
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Oral Medicine Center, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- School of Stomatology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Disease, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yunyang Liao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Oral Medicine Center, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- School of Stomatology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Disease, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shaofeng Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Oral Medicine Center, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- School of Stomatology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Disease, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Bangwei Che
- Department of Urology & Andrology, The First Affiliated of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hanghang Zhu
- School of Stomatology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Disease, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Bingbing Yang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Oral Medicine Center, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- School of Stomatology, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Disease, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Bin Shi
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Oral Medicine Center, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
15
|
Abhale K, Addepalli V, Desai S, Sanap A, Bhonde R. Effects of Mesenchymal Stem Cell-conditioned Media with Natural Immunomodulatory Agent Resveratrol on Type 1 Diabetes. Curr Drug Discov Technol 2025; 22:e080324227818. [PMID: 38468534 DOI: 10.2174/0115701638276524240305054259] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/18/2024] [Accepted: 01/29/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is a condition marked by elevated blood sugar levels and primarily recognized by the destruction of beta cells caused by an autoimmune attack, which is a significant characteristic of T1DM. Recent studies have demonstrated the regenerative potential of conditioned medium therapy. In light of this, the current research sought to assess the impact of Mesenchymal Stem Cell conditioned media (CM) and CM with resveratrol (CM+ Resveratrol) on the management of T1DM in Swiss albino mice. By leveraging and modifying existing conditioned medium therapy, this study aims to evaluate its effectiveness in treating T1DM. MATERIALS & METHODS Diabetes was induced in animals using the diabetes-inducing agent streptozotocin (STZ). The animals were then divided into five groups: Normal control, Disease Control, Resveratrol, Condition Media, and CM + Resveratrol. Treatments were given to the animals accordingly. The study period was 28 days. During this time, the animals were monitored for foodwater intake twice a week, blood glucose levels, and body weight. At the conclusion of the 28-day study period, biochemical estimations were performed for serum insulin levels, C-peptide levels, anti-inflammatory cytokines levels and pro-inflammatory cytokines levels. Additionally, histopathology of the pancreas was performed. RESULTS The test groups showed a significant decrease in blood glucose levels, an increase in Cpeptide levels, and a decrease in pro-inflammatory cytokine levels compared to the disease group. However, no statistically significant change within groups was observed in terms of serum insulin and anti-inflammatory cytokine levels. The improvement in diabetic symptoms, such as polyphagia, polydipsia, and weight loss, was observed in the treatment group, along with pancreatic regeneration, which indicated improved insulin secretion. CONCLUSION In the current investigation, we concluded that CM and CM+ Resveratrol, as natural immunomodulators, have the capacity to regenerate injured pancreatic beta cells and have antidiabetic action, together with immunomodulating impact. Nonetheless, future studies on this therapy appear to be promising.
Collapse
Affiliation(s)
- Krushna Abhale
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
| | | | - Shivani Desai
- Clinical Research and Pharmacovigilance, Serum Institute of India Pvt. Ltd., Hadapsar, Pune
| | - Avinash Sanap
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Pimpri, Pune, India
| | - Ramesh Bhonde
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Pimpri, Pune, India
| |
Collapse
|
16
|
Wei G, Zong B, He Q, Su S, Li Y, Zheng J, Qian Y, Cao P, Li Z. A Thin Polymer Layer Enables Peptide-Polycation Complexes with Ultrahigh Efficient Encapsulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2405948. [PMID: 39358966 DOI: 10.1002/smll.202405948] [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: 07/16/2024] [Revised: 08/31/2024] [Indexed: 10/04/2024]
Abstract
A monolayer encapsulation is a new opportunity for engineering a system with high drug loading, but immobilizing polymer molecules on the surface of individual peptide nanoparticles is still an ongoing challenge. Herein, an individual peptide nanoparticle encapsulation strategy is proposed via surface adsorption, in which peptide molecules undergo granulation and subsequently aggregate with polymer molecules, forming a network via electrostatic interactions. Under the water phase, surplus polymer molecules dissolve, leading to a single nanoparticle encapsulation with a core-shell structure. As expected, the dense interfacial layer on the peptide nanoparticle surface achieves a superior loading degree of up to 95.4%. What's more, once the core-shell structure is established, the peptide mass fraction in individual encapsulation always exceeds 90% even under fierce external force. Following the individual nanoparticle encapsulation, the insulin-polycation complex (InsNp@PEI) reduces the inflammation from polymer and displays an effective glycemic control in type 1 diabetes. Overall, the newly developed single surface decoration encapsulates peptides with ultrahigh efficiency and opens up the possibility for further encapsulation.
Collapse
Affiliation(s)
- Guangfei Wei
- Clinical Medical Research Center, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, 212004, China
- Affiliated Zhenjiang Integrated Hospital of Traditional Chinese and Western Medicine of Xinglin College, Nantong University, Zhenjiang, 212004, China
| | - Bin Zong
- Clinical Medical Research Center, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, 212004, China
- Affiliated Zhenjiang Integrated Hospital of Traditional Chinese and Western Medicine of Xinglin College, Nantong University, Zhenjiang, 212004, China
| | - Quan He
- Clinical Medical Research Center, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, 212004, China
- Affiliated Zhenjiang Integrated Hospital of Traditional Chinese and Western Medicine of Xinglin College, Nantong University, Zhenjiang, 212004, China
| | - Shiying Su
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Yu Li
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Jiawen Zheng
- Clinical Medical Research Center, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, 212004, China
- Affiliated Zhenjiang Integrated Hospital of Traditional Chinese and Western Medicine of Xinglin College, Nantong University, Zhenjiang, 212004, China
| | - Yuanxia Qian
- Clinical Medical Research Center, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, 212004, China
- Affiliated Zhenjiang Integrated Hospital of Traditional Chinese and Western Medicine of Xinglin College, Nantong University, Zhenjiang, 212004, China
| | - Peng Cao
- Clinical Medical Research Center, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, 212004, China
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
- Shandong Academy of Chinese Medicine, Jinan, 250014, China
| | - Zhongxing Li
- Clinical Medical Research Center, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, 212004, China
- Affiliated Zhenjiang Integrated Hospital of Traditional Chinese and Western Medicine of Xinglin College, Nantong University, Zhenjiang, 212004, China
| |
Collapse
|
17
|
Chen SD, Chu CY, Wang CB, Yang Y, Xu ZY, Qu YL, Man Y. Integrated-omics profiling unveils the disparities of host defense to ECM scaffolds during wound healing in aged individuals. Biomaterials 2024; 311:122685. [PMID: 38944969 DOI: 10.1016/j.biomaterials.2024.122685] [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: 02/05/2024] [Revised: 06/11/2024] [Accepted: 06/23/2024] [Indexed: 07/02/2024]
Abstract
Extracellular matrix (ECM) scaffold membranes have exhibited promising potential to better the outcomes of wound healing by creating a regenerative microenvironment around. However, when compared to the application in younger individuals, the performance of the same scaffold membrane in promoting re-epithelialization and collagen deposition was observed dissatisfying in aged mice. To comprehensively explore the mechanisms underlying this age-related disparity, we conducted the integrated analysis, combing single-cell RNA sequencing (scRNA-Seq) with spatial transcriptomics, and elucidated six functionally and spatially distinctive macrophage groups and lymphocytes surrounding the ECM scaffolds. Through intergroup comparative analysis and cell-cell communication, we characterized the dysfunction of Spp1+ macrophages in aged mice impeded the activation of the type Ⅱ immune response, thus inhibiting the repair ability of epidermal cells and fibroblasts around the ECM scaffolds. These findings contribute to a deeper understanding of biomaterial applications in varied physiological contexts, thereby paving the way for the development of precision-based biomaterials tailored specifically for aged individuals in future therapeutic strategies.
Collapse
Affiliation(s)
- Shuai-Dong Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chen-Yu Chu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chen-Bing Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Yang Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhao-Yu Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi-Li Qu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Man
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
18
|
Wu N, Li J, Li X, Wang R, Zhang L, Liu Z, Jiao T. 3D printed biopolymer/black phosphorus nanoscaffolds for bone implants: A review. Int J Biol Macromol 2024; 279:135227. [PMID: 39218178 DOI: 10.1016/j.ijbiomac.2024.135227] [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] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Bone implantation is one of the recognized and effective means of treating bone defects, but osteoporosis and bone tumor-related bone abnormalities have a series of problems such as susceptibility to infection, difficulty in healing, and poor therapeutic effect, which poses a great challenge to clinical medicine. Three-dimensional things may be printed using 3D printing. Researchers can feed materials through the printer layer by layer to create the desired shape for a 3D structure. It is widely employed in the healing of bone defects, and it is an improved form of additive manufacturing technology with prospective future applications. This review's objective is to provide an overview of the findings reports pertaining to 3D printing biopolymers in recent years, provide an overview of biopolymer materials and their composites with black phosphorus for 3D printing bone implants, and the characterization methods of composite materials are also summarized. In addition, summarizes 3D printing methods based on ink printing and laser printing, pointing out their special features and advantages, and provide a combination strategy of photothermal therapy and bone regeneration materials for black phosphorus-based materials. Finally, the associations between bone implant materials and immune cells, the bio-environment, as well as the 3D printing bone implants prospects are outlined.
Collapse
Affiliation(s)
- Nannan Wu
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China
| | - Jinghong Li
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China.
| | - Xinyu Li
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China
| | - Ran Wang
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China
| | - Lexin Zhang
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China
| | - Zhiwei Liu
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China
| | - Tifeng Jiao
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China.
| |
Collapse
|
19
|
Krutko M, Poling HM, Bryan AE, Sharma M, Singh A, Reza HA, Wikenheiser-Brokamp KA, Takebe T, Helmrath MA, Harris GM, Esfandiari L. Enhanced Piezoelectric Performance of PVDF-TrFE Nanofibers through Annealing for Tissue Engineering Applications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608345. [PMID: 39229142 PMCID: PMC11370437 DOI: 10.1101/2024.08.16.608345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
This study investigates bioelectric stimulation's role in tissue regeneration by enhancing the piezoelectric properties of tissue-engineered grafts using annealed poly(vinylidene fluoride-trifluoroethylene) (PVDF-TrFE) scaffolds. Annealing at temperatures of 80°C, 100°C, 120°C, and 140°C was assessed for its impact on material properties and physiological utility. Analytical techniques such as Differential Scanning Calorimetry (DSC), Fourier-Transform Infrared Spectroscopy (FTIR), and X-ray Diffraction (XRD) revealed increased crystallinity with higher annealing temperatures, peaking in β-phase content and crystallinity at 140°C. Scanning Electron Microscopy (SEM) showed that 140°C annealed scaffolds had enhanced lamellar structures, increased porosity, and maximum piezoelectric response. Mechanical tests indicated that 140°C annealing improved elastic modulus, tensile strength, and substrate stiffness, aligning these properties with physiological soft tissues. In vitro assessments in Schwann cells demonstrated favorable responses, with increased cell proliferation, contraction, and extracellular matrix attachment. Additionally, genes linked to extracellular matrix production, vascularization, and calcium signaling were upregulated. The foreign body response in C57BL/6 mice, evaluated through Hematoxylin and Eosin (H&E) and Picrosirius Red staining, showed no differences between scaffold groups, supporting the potential for future functional evaluation of the annealed group in tissue repair.
Collapse
|
20
|
Jiang H, Sun X, Wu Y, Xu J, Xiao C, Liu Q, Fang L, Liang Y, Zhou J, Wu Y, Lin Z. Contribution of Tregs to the promotion of constructive remodeling after decellularized extracellular matrix material implantation. Mater Today Bio 2024; 27:101151. [PMID: 39104900 PMCID: PMC11298607 DOI: 10.1016/j.mtbio.2024.101151] [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: 04/15/2024] [Revised: 07/02/2024] [Accepted: 07/07/2024] [Indexed: 08/07/2024] Open
Abstract
Host remodeling of decellularized extracellular matrix (dECM) material through the appropriate involvement of immune cells is essential for achieving functional organ/tissue regeneration. As many studies have focused on the role of macrophages, only few have evaluated the role of regulatory T cells (Tregs) in dECM remodeling. In this study, we used a mouse model of traumatic muscle injury to determine the role of Tregs in the constructive remodeling of vascular-derived dECM. According to the results, a certain number of Tregs could be recruited after dECM implantation. Notably, using anti-CD25 to reduce the number of Tregs recruited by the dECM was significantly detrimental to material remodeling based on a significant reduction in the number of M2 macrophages. In addition, collagen and elastic fibers, which maintain the integrity and mechanical properties of the material, rapidly degraded during the early stages of implantation. In contrast, the use of CD28-SA antibodies to increase the number of Tregs recruited by dECM promoted constructive remodeling, resulting in a decreased inflammatory response at the material edge, thinning of the surrounding fibrous connective tissue, uniform infiltration of host cells, and significantly improved tissue remodeling scores. The number of M2 macrophages increased whereas that of M1 macrophages decreased. Moreover, Treg-conditioned medium further enhanced material-induced M2 macrophage polarization in vitro. Overall, Treg is an important cell type that influences constructive remodeling of the dECM. Such findings contribute to the design of next-generation biomaterials to optimize the remodeling and regeneration of dECM materials.
Collapse
Affiliation(s)
- Hongjing Jiang
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, Guangdong, China
| | - Xuheng Sun
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
| | - Yindi Wu
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, Guangdong, China
| | - Jianyi Xu
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
| | - Cong Xiao
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
| | - Qing Liu
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
| | - Lijun Fang
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
| | - Yuanfeng Liang
- Department of Geriatrics, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510006, Guangzhou, Guangdong, China
| | - Jiahui Zhou
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, Guangdong, China
| | - Yueheng Wu
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, Guangdong, China
- Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, 528200, Foshan, Guangdong, China
| | - Zhanyi Lin
- School of Medicine, South China University of Technology, 510006, Guangzhou, Guangdong, China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, Guangdong, China
- Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, 528200, Foshan, Guangdong, China
| |
Collapse
|
21
|
Ortaleza K, Won SY, Kinney SM, Sefton MV. Aspects of the alternative host response to methacrylic acid containing biomaterials. J Biomed Mater Res A 2024; 112:1276-1285. [PMID: 38053493 DOI: 10.1002/jbm.a.37652] [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: 08/03/2023] [Revised: 10/05/2023] [Accepted: 11/20/2023] [Indexed: 12/07/2023]
Abstract
Methacrylic acid (MAA)-based biomaterials promote a vascularized host response without the addition of exogenous factors such as cells or growth factors. We presume that materials containing MAA favor an alternative foreign body response, rather than the conventional fibrotic response. Here, we characterize selected aspects of the response to two different forms of MAA-a coating, which can be used to prevascularize the subcutaneous tissue for subsequent therapeutic cell delivery or an injectable hydrogel, which can be used to vascularize and deliver cells simultaneously. We show that the MAA-coating quickly vascularized the subcutaneous space compared to an uncoated silicone tube, and after 14 days of prevascularization, the tissue surrounding the MAA-coated tube presented fewer immune cells than the uncoated control. We also compared the host response to a MAA-PEG (polyethylene glycol) hydrogel at day 1, with pancreatic islets in immune-compromised SCID/bg mice and immune-competent Balb/c mice. The Balb/c mouse presented a more inflammatory response with increased IFN-γ production as compared to the SCID/bg. Together with previously published data, this work contributes to a further understanding of tissue responses to a biomaterial in different forms as used for cell delivery.
Collapse
Affiliation(s)
- Krystal Ortaleza
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - So-Yoon Won
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Sean M Kinney
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Michael V Sefton
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Wang Z, Shi H, Silveira PA, Mithieux SM, Wong WC, Liu L, Pham NTH, Hawkett BS, Wang Y, Weiss AS. Tropoelastin modulates systemic and local tissue responses to enhance wound healing. Acta Biomater 2024; 184:54-67. [PMID: 38871204 DOI: 10.1016/j.actbio.2024.06.009] [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: 12/11/2023] [Revised: 05/13/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Wound healing is facilitated by biomaterials-based grafts and substantially impacted by orchestrated inflammatory responses that are essential to the normal repair process. Tropoelastin (TE) based materials are known to shorten the period for wound repair but the mechanism of anti-inflammatory performance is not known. To explore this, we compared the performance of the gold standard Integra Dermal Regeneration Template (Integra), polyglycerol sebacate (PGS), and TE blended with PGS, in a murine full-thickness cutaneous wound healing study. Systemically, blending with TE favorably increased the F4/80+ macrophage population by day 7 in the spleen and contemporaneously induced elevated plasma levels of anti-inflammatory IL-10. In contrast, the PGS graft without TE prompted prolonged inflammation, as evidenced by splenomegaly and greater splenic granulocyte and monocyte fractions at day 14. Locally, the inclusion of TE in the graft led to increased anti-inflammatory M2 macrophages and CD4+T cells at the wound site, and a rise in Foxp3+ regulatory T cells in the wound bed by day 7. We conclude that the TE-incorporated skin graft delivers a pro-healing environment by modulating systemic and local tissue responses. STATEMENT OF SIGNIFICANCE: Tropoelastin (TE) has shown significant benefits in promoting the repair and regeneration of damaged human tissues. In this study, we show that TE promotes an anti-inflammatory environment that facilitates cutaneous wound healing. In a mouse model, we find that inserting a TE-containing material into a full-thickness wound results in defined, pro-healing local and systemic tissue responses. These findings advance our understanding of TE's restorative value in tissue engineering and regenerative medicine, and pave the way for clinical applications.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Life and Environmental Sciences, the University of Sydney, NSW 2006, Australia; Charles Perkins Centre, the University of Sydney, NSW 2006, Australia
| | - Huaikai Shi
- Burns Research and Reconstructive Surgery, Anzac Research Institute, NSW 2139, Australia; Asbestos and Dust Disease Research Institute, Concord Hospital, Sydney, NSW 2139, Australia
| | - Pablo A Silveira
- Dendritic Cell Group, ANZAC Research Institute, Concord Hospital, Sydney, NSW 2139, Australia
| | - Suzanne M Mithieux
- School of Life and Environmental Sciences, the University of Sydney, NSW 2006, Australia; Charles Perkins Centre, the University of Sydney, NSW 2006, Australia
| | - Wai Cheng Wong
- Charles Perkins Centre, the University of Sydney, NSW 2006, Australia
| | - Linyang Liu
- School of Life and Environmental Sciences, the University of Sydney, NSW 2006, Australia; Charles Perkins Centre, the University of Sydney, NSW 2006, Australia
| | - Nguyen T H Pham
- Key Centre for Polymers and Colloids, School of Chemistry, the University of Sydney, NSW 2006, Australia
| | - Brian S Hawkett
- Key Centre for Polymers and Colloids, School of Chemistry, the University of Sydney, NSW 2006, Australia
| | - Yiwei Wang
- Burns Research and Reconstructive Surgery, Anzac Research Institute, NSW 2139, Australia; Jiangsu Provincial Engineering Research Centre of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| | - Anthony S Weiss
- School of Life and Environmental Sciences, the University of Sydney, NSW 2006, Australia; Charles Perkins Centre, the University of Sydney, NSW 2006, Australia; The University of Sydney Nano Institute, the University of Sydney, NSW 2006, Australia.
| |
Collapse
|
23
|
Farzamfar S, Garcia LM, Rahmani M, Bolduc S. Navigating the Immunological Crossroads: Mesenchymal Stem/Stromal Cells as Architects of Inflammatory Harmony in Tissue-Engineered Constructs. Bioengineering (Basel) 2024; 11:494. [PMID: 38790361 PMCID: PMC11118848 DOI: 10.3390/bioengineering11050494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
In the dynamic landscape of tissue engineering, the integration of tissue-engineered constructs (TECs) faces a dual challenge-initiating beneficial inflammation for regeneration while avoiding the perils of prolonged immune activation. As TECs encounter the immediate reaction of the immune system upon implantation, the unique immunomodulatory properties of mesenchymal stem/stromal cells (MSCs) emerge as key navigators. Harnessing the paracrine effects of MSCs, researchers aim to craft a localized microenvironment that not only enhances TEC integration but also holds therapeutic promise for inflammatory-driven pathologies. This review unravels the latest advancements, applications, obstacles, and future prospects surrounding the strategic alliance between MSCs and TECs, shedding light on the immunological symphony that guides the course of regenerative medicine.
Collapse
Affiliation(s)
- Saeed Farzamfar
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
| | - Luciana Melo Garcia
- Department of Medicine, Université Laval, Québec, QC G1V 0A6, Canada;
- Hematology-Oncology Service, CHU de Québec—Université Laval, Québec, QC G1V 0A6, Canada
| | - Mahya Rahmani
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
| | - Stephane Bolduc
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
24
|
Aarthy M, Hemalatha T, Suryalakshmi P, Vinoth V, Mercyjayapriya J, Shanmugam G, Ayyadurai N. Biomimetic design of fibril-forming non-immunogenic collagen like proteins for tissue engineering. Int J Biol Macromol 2024; 266:130999. [PMID: 38521303 DOI: 10.1016/j.ijbiomac.2024.130999] [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: 11/24/2023] [Revised: 02/29/2024] [Accepted: 03/17/2024] [Indexed: 03/25/2024]
Abstract
Collagen, a key component of extracellular matrix serves as a linchpin for maintaining structural integrity and functional resilience. Concerns over purity and immunogenicity of animal-derived collagens have spurred efforts to develop synthetic collagen-based biomaterials. Despite several collagen mimics, there remains limited exploration of non-immunogenic biomaterials with the capacity for effective self-assembly. To combat the lacuna, collagen like protein (CLP) variants were rationally designed and recombinantly expressed, incorporating human telopeptide sequences (CLP-N and CLP-NC) and bioactive binding sites (CLP-NB). Circular dichroism analyses of the variants confirmed the triple helical conformation, with variations in thermal stability and conformation attributed to the presence of telopeptides at one or both ends of CLP. The variants had propensity to form oligomers, setting the stage for fibrillogenesis. The CLP variants were biocompatible, hemocompatible and supported cell proliferation and migration, particularly CLP-NB with integrin-binding sites. Gene expression indicated a lack of significant upregulation of inflammatory markers, highlighting the non-immunogenic nature of these variants. Lyophilized CLP scaffolds maintained their triple-helical structure and offered favorable biomaterial characteristics. These results accentuate the potential of designed CLP variants in tissue engineering, regenerative medicine and industrial sectors, supporting the development of biocompatible scaffolds and implants for therapeutic and cosmetic purposes.
Collapse
Affiliation(s)
- Mayilvahanan Aarthy
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) - Central Leather Research Institute (CLRI), Chennai 600020, India
| | - Thiagarajan Hemalatha
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) - Central Leather Research Institute (CLRI), Chennai 600020, India
| | - Pandurangan Suryalakshmi
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) - Central Leather Research Institute (CLRI), Chennai 600020, India
| | - Vetrivel Vinoth
- Department of Organic and Bioorganic Chemistry, CSIR-CLRI, Chennai 600020, India
| | - Jebakumar Mercyjayapriya
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) - Central Leather Research Institute (CLRI), Chennai 600020, India
| | - Ganesh Shanmugam
- Department of Organic and Bioorganic Chemistry, CSIR-CLRI, Chennai 600020, India
| | - Niraikulam Ayyadurai
- Department of Biochemistry and Biotechnology, Council of Scientific and Industrial Research (CSIR) - Central Leather Research Institute (CLRI), Chennai 600020, India.
| |
Collapse
|
25
|
Zareein A, Mahmoudi M, Jadhav SS, Wilmore J, Wu Y. Biomaterial engineering strategies for B cell immunity modulations. Biomater Sci 2024; 12:1981-2006. [PMID: 38456305 PMCID: PMC11019864 DOI: 10.1039/d3bm01841e] [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: 11/11/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
B cell immunity has a penetrating effect on human health and diseases. Therapeutics aiming to modulate B cell immunity have achieved remarkable success in combating infections, autoimmunity, and malignancies. However, current treatments still face significant limitations in generating effective long-lasting therapeutic B cell responses for many conditions. As the understanding of B cell biology has deepened in recent years, clearer regulation networks for B cell differentiation and antibody production have emerged, presenting opportunities to overcome current difficulties and realize the full therapeutic potential of B cell immunity. Biomaterial platforms have been developed to leverage these emerging concepts to augment therapeutic humoral immunity by facilitating immunogenic reagent trafficking, regulating T cell responses, and modulating the immune microenvironment. Moreover, biomaterial engineering tools have also advanced our understanding of B cell biology, further expediting the development of novel therapeutics. In this review, we will introduce the general concept of B cell immunobiology and highlight key biomaterial engineering strategies in the areas including B cell targeted antigen delivery, sustained B cell antigen delivery, antigen engineering, T cell help optimization, and B cell suppression. We will also discuss our perspective on future biomaterial engineering opportunities to leverage humoral immunity for therapeutics.
Collapse
Affiliation(s)
- Ali Zareein
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Mina Mahmoudi
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Shruti Sunil Jadhav
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
| | - Joel Wilmore
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
26
|
Giri PS, Rath SN. Macrophage Polarization Dynamics in Biomaterials: Implications for in Vitro Wound Healing. ACS APPLIED BIO MATERIALS 2024; 7:2413-2422. [PMID: 38536097 DOI: 10.1021/acsabm.4c00066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The interaction between biomaterials and the immune system plays a pivotal role in determining the success or failure of implantable devices. Macrophages, as key orchestrators of immune responses, exhibit diverse reactions that influence tissue integration or lead to implant failure. This study focuses on unraveling the intricate relationship between macrophage phenotypes and biomaterials, specifically hydrogels, by employing THP-1 cells as a model. Through a comprehensive investigation using polysaccharide, polymer, and protein-based hydrogels, our research sheds light on how the properties of hydrogels influence macrophage polarization. Phenotypic observations, biochemical assays, surface marker expression, and gene expression profiles collectively demonstrate the differential macrophage polarization abilities of polysaccharide-, polymer-, and protein-based hydrogels. Moreover, our indirect coculture studies reveal that hydrogels fostering M2 polarization exhibit exceptional wound-healing capabilities. These findings highlight the crucial role of the hydrogel microenvironment in adjusting macrophage polarization, offering a fresh avenue for refining biomaterials to bolster advantageous immune responses and improve tissue integration. This research contributes valuable insights for designing biomaterials with tailored properties that can guide macrophage behavior, ultimately improving the overall success of implantable devices.
Collapse
Affiliation(s)
- Pravin Shankar Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284 Telangana, India
| | - Subha Narayan Rath
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284 Telangana, India
| |
Collapse
|
27
|
Du Y, Deng T, Cheng Y, Zhao Q, Xia H, Ji Y, Zhang Y, He Q. Enhancing Bone Regeneration through CDC20-Loaded ZIF-8 Nanoparticles Wrapped in Erythrocyte Membranes with Targeting Aptamer. Adv Healthc Mater 2024; 13:e2302725. [PMID: 38030141 DOI: 10.1002/adhm.202302725] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/15/2023] [Indexed: 12/01/2023]
Abstract
In the context of bone regeneration, nanoparticles harboring osteogenic factors have emerged as pivotal agents for modulating the differentiation fate of stem cells. However, persistent challenges surrounding biocompatibility, loading efficiency, and precise targeting ability warrant innovative solution. In this study, a novel nanoparticle platform founded upon the zeolitic imidazolate framework-8 (ZIF-8) is introduced. This new design, CDC20@ZIF-8@eM-Apt, involves the envelopment of ZIF-8 within an erythrocyte membrane (eM) cloak, and is coupled with a targeting aptamer. ZIF-8, distinguished by its porosity, biocompatibility, and robust cargo transport capabilities, constitutes the core framework. Cell division cycle protein 20 homolog (CDC20) is illuminated as a new target in bone regeneration. The eM plays a dual role in maintaining nanoparticle stability and facilitating fusion with target cell membranes, while the aptamer orchestrates the specific recruitment of bone marrow mesenchymal stem cells (BMSCs) within bone defect sites. Significantly, CDC20@ZIF-8@eM-Apt amplifies osteogenic differentiation of BMSCs via the inhibition of NF-κB p65, and concurrently catalyzes bone regeneration in two bone defect models. Consequently, CDC20@ZIF-8@eM-Apt introduces a pioneering strategy for tackling bone defects and associated maladies, opening novel avenues in therapeutic intervention.
Collapse
Affiliation(s)
- Yangge Du
- 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, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Tian Deng
- 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, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Yihong Cheng
- 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, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Qin Zhao
- 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, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Haibin Xia
- 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, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Yaoting Ji
- 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, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, 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, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Qing 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, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
28
|
Fateh S, Alromaihi RA, Ghaemmaghami AM, Alexander MR. Unlocking Bio-Instructive Polymers: A Novel Multi-Well Screening Platform Based on Secretome Sampling. Bio Protoc 2024; 14:e4939. [PMID: 38405080 PMCID: PMC10883890 DOI: 10.21769/bioprotoc.4939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/01/2024] [Accepted: 01/05/2024] [Indexed: 02/27/2024] Open
Abstract
Biomaterials are designed to interact with biological systems to replace, support, enhance, or monitor their function. However, there are challenges associated with traditional biomaterials' development due to the lack of underlying theory governing cell response to materials' chemistry. This leads to the time-consuming process of testing different materials plus the adverse reactions in the body such as cytotoxicity and foreign body response. High-throughput screening (HTS) offers a solution to these challenges by enabling rapid and simultaneous testing of a large number of materials to determine their bio-interactions and biocompatibility. Secreted proteins regulate many physiological functions and determine the success of implanted biomaterials through directing cell behaviour. However, the majority of biomaterials' HTS platforms are suitable for microscopic analyses of cell behaviour and not for investigating non-adherent cells or measuring cell secretions. Here, we describe a multi-well platform adaptable to robotic printing of polymers and suitable for secretome profiling of both adherent and non-adherent cells. We detail the platform's development steps, encompassing the preparation of individual cell culture chambers, polymer printing, and the culture environment, as well as examples to demonstrate surface chemical characterisation and biological assessments of secreted mediators. Such platforms will no doubt facilitate the discovery of novel biomaterials and broaden their scope by adapting wider arrays of cell types and incorporating assessments of both secretome and cell-bound interactions. Key features • Detailed protocols for preparation of substrate for contact printing of acrylate-based polymers including O2 plasma etching, functionalisation process, and Poly(2-hydroxyethyl methacrylate) (pHEMA) dip coating. • Preparations of 7 mm × 7 mm polymers employing pin printing system. • Provision of confined area for each polymer using ProPlate® multi-well chambers. • Compatibility of this platform was validated using adherent cells [primary human monocyte-derived macrophages (MDMs)) and non-adherent cells (primary human monocyte-derived dendritic cells (moDCs)]. • Examples of the adaptability of the platform for secretome analysis including five different cytokines using enzyme-linked immunosorbent assay (ELISA, DuoSet®). Graphical overview.
Collapse
Affiliation(s)
- Shirin Fateh
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | | | | | | |
Collapse
|
29
|
Xu Y, Saiding Q, Zhou X, Wang J, Cui W, Chen X. Electrospun fiber-based immune engineering in regenerative medicine. SMART MEDICINE 2024; 3:e20230034. [PMID: 39188511 PMCID: PMC11235953 DOI: 10.1002/smmd.20230034] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/26/2024] [Indexed: 08/28/2024]
Abstract
Immune engineering, a burgeoning field within regenerative medicine, involves a spectrum of strategies to optimize the intricate interplay between tissue regenerative biomaterials and the host tissue. These strategies are applied across different types of biomaterials and various disease models, which encompasses finely modulating the immune response at the levels of immune cells and factors, aiming to mitigate adverse effects like fibrosis and persistent inflammation that may arise at the injury site and consequently promote tissue regeneration. With the continuous progress in electrospinning technology, the immunoregulatory capabilities of electrospun fibers have gained substantial attention over the years. Electrospun fibers, with their extracellular matrix-like characteristics, high surface-area-to-volume ratio, and reliable pharmaceutical compound capacity, have emerged as key players among tissue engineering materials. This review specifically focuses on the role of electrospun fiber-based immune engineering, emphasizing their unique design strategies. Notably, electrospinning actively engages in immune engineering by modulating immune responses through four essential strategies: (i) surface modification, (ii) drug loading, (iii) physicochemical parameters, and (iv) biological grafting. This review presents a comprehensive overview of the intricate mechanisms of the immune system in injured tissues while unveiling the key strategies adopted by electrospun fibers to orchestrate immune regulation. Furthermore, the review explores the current developmental trends and limitations concerning the immunoregulatory function of electrospun fibers, aiming to drive the advancements in electrospun fiber-based immune engineering to its full potential.
Collapse
Affiliation(s)
- Yiru Xu
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| | - Qimanguli Saiding
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue Zhou
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| | - Juan Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xinliang Chen
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| |
Collapse
|
30
|
Shi Y, Tao W, Yang W, Wang L, Qiu Z, Qu X, Dang J, He J, Fan H. Calcium phosphate coating enhances osteointegration of melt electrowritten scaffold by regulating macrophage polarization. J Nanobiotechnology 2024; 22:47. [PMID: 38297240 PMCID: PMC10829397 DOI: 10.1186/s12951-024-02310-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/26/2024] [Indexed: 02/02/2024] Open
Abstract
The osteoimmune microenvironment induced by implants plays a significant role in bone regeneration. It is essential to efficiently and timely switch the macrophage phenotype from M1 to M2 for optimal bone healing. This study examined the impact of a calcium phosphate (CaP) coating on the physiochemical properties of highly ordered polycaprolactone (PCL) scaffolds fabricated using melt electrowritten (MEW). Additionally, it investigated the influence of these scaffolds on macrophage polarization and their immunomodulation on osteogenesis. The results revealed that the CaP coated PCL scaffold exhibited a rougher surface topography and higher hydrophilicity in comparison to the PCL scaffold without coating. Besides, the surface morphology of the coating and the release of Ca2+ from the CaP coating were crucial in regulating the transition of macrophages from M1 to M2 phenotypes. They might activate the PI3K/AKT and cAMP-PKA pathways, respectively, to facilitate M2 polarization. In addition, the osteoimmune microenvironment induced by CaP coated PCL could not only enhance the osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) in vitro but also promote the bone regeneration in vivo. Taken together, the CaP coating can be employed to control the phenotypic switching of macrophages, thereby creating a beneficial immunomodulatory microenvironment that promotes bone regeneration.
Collapse
Affiliation(s)
- Yubo Shi
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Weidong Tao
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wenjing Yang
- Xijing 986 Hospital Department, The Fourth Military Medical University, Xi'an, China
| | - Lei Wang
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhennan Qiu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoli Qu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Dang
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an, China
| | - Hongbin Fan
- Department of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
31
|
Xie X, Xu J, Lin J, Chen L, Ding D, Hu Y, Han K, Li C, Wang F, Zhao J, Wang L. Micro-nano hierarchical scaffold providing temporal-matched biological constraints for tendon reconstruction. Biofabrication 2023; 16:015018. [PMID: 38100814 DOI: 10.1088/1758-5090/ad1608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 12/15/2023] [Indexed: 12/17/2023]
Abstract
Due to the limitations of tendon biology, high-quality tendon repair remains a clinical and scientific challenge. Here, a micro-nano hierarchical scaffold is developed to promote orderly tendon regeneration by providing temporal-matched biological constraints. In short, fibrin (Fb), which provides biological constraints, is loaded into poly (DL-lactide-co-glycolide) nanoyarns with suitable degradation cycles (Fb-loaded nanofiber yarns (Fb-NY)). Then further combined with braiding technology, temporary chemotactic Fb scaffolds with tendon extracellular matrix-like structures are obtained to initiate the regeneration process. At the early stage of healing (2 w), the regeneration microenvironment is regulated (inducing M2 macrophages and restoring the early blood supply necessary for healing) by Fb, and the alignment of cells and collagen is induced by nanoyarn. At the late healing stage (8 w), with the degradation of Fb-NY, non-functional vascular regression occurs, and the newborn tissues gradually undergo load-bearing remodeling, restoring the anvascularous and ordered structure of the tendon. In summary, the proposed repair strategy provides temporal-matched biological constraints, offering a potential pathway to reconstruct the ordered structure and function of tendons.
Collapse
Affiliation(s)
- Xiaojing Xie
- Key Laboratory of Textile Science & Technology of Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, People's Republic of China
| | - Junjie Xu
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
| | - Jing Lin
- Key Laboratory of Textile Science & Technology of Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, People's Republic of China
| | - Liang Chen
- National Institutes for Food and Drug Control, Beijing 102629, People's Republic of China
| | - Danzhi Ding
- Key Laboratory of Textile Science & Technology of Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, People's Republic of China
| | - Yage Hu
- Key Laboratory of Textile Science & Technology of Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, People's Republic of China
| | - Kang Han
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
| | - Chaojing Li
- Key Laboratory of Textile Science & Technology of Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, People's Republic of China
| | - Fujun Wang
- Key Laboratory of Textile Science & Technology of Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, People's Republic of China
| | - Jinzhong Zhao
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
| | - Lu Wang
- Key Laboratory of Textile Science & Technology of Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, People's Republic of China
| |
Collapse
|
32
|
Sugiura Y, Yamada E, Horie M. Fabrication of hydrophilic polymer-hybrid octacalcium phosphate blocks under wet condition based on cement setting reactions. J Mech Behav Biomed Mater 2023; 148:106226. [PMID: 37952506 DOI: 10.1016/j.jmbbm.2023.106226] [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: 09/22/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 11/14/2023]
Abstract
Bioceramics, while offering excellent biocompatibility, are often compromised by their fragility and brittleness, especially under wet conditions. Even though recent hybrid processes combining biocompatible polymers and bioceramics have shown promise, complete mitigation of these challenges remains elusive. In this research, a biomimetic process was employed to mimic the structure of biological bone tissue. This led to the development of block materials composed of octacalcium phosphate (OCP) and sodium polyacrylic acid (PAA-Na) that display flexibility and resilience in wet conditions. Adjusting the PAA-Na concentration enabled the OCP-PAA-Na blocks to demonstrate superior mechanical strength when dry and increased flexibility when wet. Notably, these blocks expanded in aqueous solutions while preserving their structure, making them ideal for oral surgeries by preventing issues like blood flooding from implanted areas.
Collapse
Affiliation(s)
- Yuki Sugiura
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14, Hayashi-cho, Takamatsu, Kagawa, 761-0395, Japan; Research Planning Office, Headquarter of Department of Life and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1, Umezono, Tsukuba, Ibaragi, 305-8560, Japan.
| | - Etsuko Yamada
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14, Hayashi-cho, Takamatsu, Kagawa, 761-0395, Japan
| | - Masanori Horie
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14, Hayashi-cho, Takamatsu, Kagawa, 761-0395, Japan
| |
Collapse
|
33
|
Darzi S, Alappadan J, Paul K, Mazdumder P, Rosamilia A, Truong YB, Gargett C, Werkmeister J, Mukherjee S. Immunobiology of foreign body response to composite PLACL/gelatin electrospun nanofiber meshes with mesenchymal stem/stromal cells in a mouse model: Implications in pelvic floor tissue engineering and regeneration. BIOMATERIALS ADVANCES 2023; 155:213669. [PMID: 37980818 DOI: 10.1016/j.bioadv.2023.213669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 11/21/2023]
Abstract
Pelvic Organ Prolapse (POP) is a common gynaecological disorder where pelvic organs protrude into the vagina. While transvaginal mesh surgery using non-degradable polymers was a commonly accepted treatment for POP, it has been associated with high rates of adverse events such as mesh erosion, exposure and inflammation due to serious foreign body response and therefore banned from clinical use after regulatory mandates. This study proposes a tissue engineering strategy using uterine endometrium-derived mesenchymal stem/stromal cells (eMSC) delivered with degradable poly L-lactic acid-co-poly ε-caprolactone (PLACL) and gelatin (G) in form of a composite electrospun nanofibrous mesh (P + G nanomesh) and evaluates the immunomodulatory mechanism at the material interfaces. The study highlights the critical acute and chronic inflammatory markers along with remodelling factors that determine the mesh surgery outcome. We hypothesise that such a bioengineered construct enhances mesh integration and mitigates the Foreign Body Response (FBR) at the host interface associated with mesh complications. Our results show that eMSC-based nanomesh significantly increased 7 genes associated with ECM synthesis and cell adhesion including, Itgb1, Itgb2, Vcam1, Cd44, Cdh2, Tgfb1, Tgfbr1, 6 genes related to angiogenesis including Ang1, Ang2, Vegfa, Pdgfa, Serpin1, Cxcl12, and 5 genes associated with collagen remodelling Col1a1, Col3a1, Col6a1, Col6a2, Col4a5 at six weeks post-implantation. Our findings suggest that cell-based tissue-engineered constructs potentially mitigate the FBR response elicited by biomaterial implants. From a clinical perspective, this construct provides an alternative to current inadequacies in surgical outcomes by modulating the immune response, inducing angiogenesis and ECM synthesis during the acute and chronic phases of the FBR.
Collapse
Affiliation(s)
- Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Janet Alappadan
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Kallyanashis Paul
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Permita Mazdumder
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Anna Rosamilia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia; Pelvic Floor Disorders Unit, Monash Health, Clayton, VIC 3168, Australia
| | | | - Caroline Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Jerome Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Shayanti Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
34
|
Liu Q, Wei F, Coathup M, Shen W, Wu D. Effect of Porosity and Pore Shape on the Mechanical and Biological Properties of Additively Manufactured Bone Scaffolds. Adv Healthc Mater 2023; 12:e2301111. [PMID: 37689976 DOI: 10.1002/adhm.202301111] [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: 04/08/2023] [Revised: 08/02/2023] [Indexed: 09/11/2023]
Abstract
This study investigates the effect of porosity and pore shape on the biological and mechanical behavior of additively manufactured scaffolds for bone tissue engineering (BTE). Polylactic acid scaffolds with varying porosity levels (15-78%) and pore shapes, including regular (rectangular pores), gyroid, and diamond (triply periodic minimal surfaces) structures, are fabricated by fused filament fabrication. Murine-derived macrophages and human bone marrow-derived mesenchymal stromal cells (hBMSCs) are seeded onto the scaffolds. The compressive behavior and surface morphology of the scaffolds are characterized. The results show that scaffolds with 15%, 30%, and 45% porosity display the highest rate of macrophage and hBMSC growth. Gyroid and diamond scaffolds exhibit a higher rate of macrophage proliferation, while diamond scaffolds exhibit a higher rate of hBMSC proliferation. Additionally, gyroid and diamond scaffolds exhibit better compressive behavior compared to regular scaffolds. Of particular note, diamond scaffolds have the highest compressive modulus and strength. Surface morphology characterization indicates that the surface roughness of diamond and gyroid scaffolds is greater than that of regular scaffolds at the same porosity level, which is beneficial for cell attachment and proliferation. This study provides valuable insights into porosity and pore shape selection for additively manufactured scaffolds in BTE.
Collapse
Affiliation(s)
- Qingyang Liu
- Department of Mechanical and Aerospace Engineering, College of Engineering and Computer Science, University of Central Florida, Orlando, FL, 32816, USA
| | - Fei Wei
- Biionix Cluster, University of Central Florida, Orlando, FL, 32827, USA
| | - Melanie Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL, 32827, USA
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| | - Wen Shen
- Department of Mechanical and Aerospace Engineering, College of Engineering and Computer Science, University of Central Florida, Orlando, FL, 32816, USA
- Biionix Cluster, University of Central Florida, Orlando, FL, 32827, USA
| | - Dazhong Wu
- Department of Mechanical and Aerospace Engineering, College of Engineering and Computer Science, University of Central Florida, Orlando, FL, 32816, USA
- Biionix Cluster, University of Central Florida, Orlando, FL, 32827, USA
| |
Collapse
|
35
|
Lowe KO, Tanase CE, Maghami S, Fisher LE, Ghaemmaghami AM. Inflammatory Network of Liver Fibrosis and How It Can Be Targeted Therapeutically. IMMUNO 2023; 3:375-408. [DOI: 10.3390/immuno3040023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Liver fibrosis is a complex, dynamic process associated with a broad spectrum of chronic liver diseases and acute liver failure, characterised by the dysregulated intrahepatic production of extracellular matrix proteins replacing functional liver cells with scar tissue. Fibrosis progresses due to an interrelated cycle of hepatocellular injury, triggering a persistent wound-healing response. The accumulation of scar tissue and chronic inflammation can eventually lead to cirrhosis and hepatocellular carcinoma. Currently, no therapies exist to directly treat or reverse liver fibrosis; hence, it remains a substantial global disease burden. A better understanding of the intricate inflammatory network that drives the initiation and maintenance of liver fibrosis to enable the rationale design of new intervention strategies is required. This review clarifies the most current understanding of the hepatic fibrosis cellular network with a focus on the role of regulatory T cells, and a possible trajectory for T cell immunotherapy in fibrosis treatment. Despite good progress in elucidating the role of the immune system in liver fibrosis, future work to better define the function of different immune cells and their mediators at different fibrotic stages is needed, which will enhance the development of new therapies.
Collapse
Affiliation(s)
- Kirstin O. Lowe
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | | - Susan Maghami
- Hull York Medical School, University of York, York YO10 5DD, UK
| | - Leanne E. Fisher
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | |
Collapse
|
36
|
Arciola CR, Ravaioli S, Mirzaei R, Dolzani P, Montanaro L, Daglia M, Campoccia D. Biofilms in Periprosthetic Orthopedic Infections Seen through the Eyes of Neutrophils: How Can We Help Neutrophils? Int J Mol Sci 2023; 24:16669. [PMID: 38068991 PMCID: PMC10706149 DOI: 10.3390/ijms242316669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Despite advancements in our knowledge of neutrophil responses to planktonic bacteria during acute inflammation, much remains to be elucidated on how neutrophils deal with bacterial biofilms in implant infections. Further complexity transpires from the emerging findings on the role that biomaterials play in conditioning bacterial adhesion, the variety of biofilm matrices, and the insidious measures that biofilm bacteria devise against neutrophils. Thus, grasping the entirety of neutrophil-biofilm interactions occurring in periprosthetic tissues is a difficult goal. The bactericidal weapons of neutrophils consist of the following: ready-to-use antibacterial proteins and enzymes stored in granules; NADPH oxidase-derived reactive oxygen species (ROS); and net-like structures of DNA, histones, and granule proteins, which neutrophils extrude to extracellularly trap pathogens (the so-called NETs: an allusive acronym for "neutrophil extracellular traps"). Neutrophils are bactericidal (and therefore defensive) cells endowed with a rich offensive armamentarium through which, if frustrated in their attempts to engulf and phagocytose biofilms, they can trigger the destruction of periprosthetic bone. This study speculates on how neutrophils interact with biofilms in the dramatic scenario of implant infections, also considering the implications of this interaction in view of the design of new therapeutic strategies and functionalized biomaterials, to help neutrophils in their arduous task of managing biofilms.
Collapse
Affiliation(s)
- Carla Renata Arciola
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Stefano Ravaioli
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Paolo Dolzani
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
| | - Lucio Montanaro
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy;
| | - Davide Campoccia
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| |
Collapse
|
37
|
Zheng W, Wu D, Zhang Y, Luo Y, Yang L, Xu X, Luo F. Multifunctional modifications of polyetheretherketone implants for bone repair: A comprehensive review. BIOMATERIALS ADVANCES 2023; 154:213607. [PMID: 37651963 DOI: 10.1016/j.bioadv.2023.213607] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023]
Abstract
Polyetheretherketone (PEEK) has emerged as a highly promising orthopedic implantation material due to its elastic modulus which is comparable to that of natural bone. This polymer exhibits impressive properties for bone implantation such as corrosion resistance, fatigue resistance, self-lubrication and chemical stability. Significantly, compared to metal-based implants, PEEK implants have mechanical properties that are closer to natural bone, which can mitigate the "stress shielding" effect in bone implantation. Nevertheless, PEEK is incapable of inducing osteogenesis due to its bio-inert molecular structure, thereby hindering the osseointegration process. To optimize the clinical application of PEEK, researchers have been working on promoting its bioactivity and endowing this polymer with beneficial properties, such as antibacterial, anti-inflammatory, anti-tumor, and angiogenesis-promoting capabilities. Considering the significant growth of research on PEEK implants over the past 5 years, this review aims to present a timely update on PEEK's modification methods. By highlighting the latest advancements in PEEK modification, we hope to provide guidance and inspiration for researchers in developing the next generation bone implants and optimizing their clinical applications.
Collapse
Affiliation(s)
- Wenzhuo Zheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Dongxu Wu
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yaowen Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yankun Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Lei Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiangrui Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Feng Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China; Department of Prosthodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
38
|
Ivanova E, Fayzullin A, Minaev N, Dolganova I, Serejnikova N, Gafarova E, Tokarev M, Minaeva E, Aleksandrova P, Reshetov I, Timashev P, Shekhter A. Surface Topography of PLA Implants Defines the Outcome of Foreign Body Reaction: An In Vivo Study. Polymers (Basel) 2023; 15:4119. [PMID: 37896364 PMCID: PMC10610271 DOI: 10.3390/polym15204119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/19/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
The formation of a dense fibrous capsule around the foreign body and its contracture is the most common complication of biomaterial implantation. The aim of our research is to find out how the surface of the implant influences the inflammatory and fibrotic reactions in the surrounding tissues. We made three types of implants with a remote surface topography formed of polylactide granules with different diameters: large (100-200 µm), medium (56-100 µm) and small (1-56 µm). We placed these implants in skin pockets in the ears of six chinchilla rabbits. We explanted the implants on the 7th, 14th, 30th and 60th days and performed optical coherence tomography, and histological, immunohistochemical and morphometric studies. We examined 72 samples and compared the composition of immune cell infiltration, vascularization, the thickness of the peri-implant tissues, the severity of fibrotic processes and α-SMA expression in myofibroblasts. We analyzed the scattering coefficient of tissue layers on OCT scans. We found that implants made from large granules induced a milder inflammatory process and slower formation of a connective tissue capsule around the foreign body. Our results prove the importance of assessing the surface texture in order to avoid the formation of capsular contracture after implantation.
Collapse
Affiliation(s)
- Elena Ivanova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia; (E.I.); (A.F.); (N.S.); (E.G.); (M.T.); (P.T.)
- B.V. Petrovsky Russian Research Center of Surgery, 2 Abrikosovskiy Lane, Moscow 119991, Russia
| | - Alexey Fayzullin
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia; (E.I.); (A.F.); (N.S.); (E.G.); (M.T.); (P.T.)
| | - Nikita Minaev
- Institute of Photon Technologies of FSRC “Crystallography and Photonics” RAS, Troitsk, Moscow 108840, Russia; (N.M.); (E.M.)
| | - Irina Dolganova
- Osipyan Institute of Solid State Physics of the Russian Academy of Sciences, 2 Osipyan St., Chernogolovka 142432, Russia;
| | - Natalia Serejnikova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia; (E.I.); (A.F.); (N.S.); (E.G.); (M.T.); (P.T.)
| | - Elvira Gafarova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia; (E.I.); (A.F.); (N.S.); (E.G.); (M.T.); (P.T.)
| | - Mark Tokarev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia; (E.I.); (A.F.); (N.S.); (E.G.); (M.T.); (P.T.)
| | - Ekaterina Minaeva
- Institute of Photon Technologies of FSRC “Crystallography and Photonics” RAS, Troitsk, Moscow 108840, Russia; (N.M.); (E.M.)
| | - Polina Aleksandrova
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilova St., Moscow 119991, Russia;
| | - Igor Reshetov
- L.L. Levshin Institute of Cluster Oncology, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia;
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia; (E.I.); (A.F.); (N.S.); (E.G.); (M.T.); (P.T.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
| | - Anatoly Shekhter
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia; (E.I.); (A.F.); (N.S.); (E.G.); (M.T.); (P.T.)
| |
Collapse
|
39
|
Protsepko O, Voisard P, Kuhn C, Maccagno A, Dannecker C, Jeschke U, Pauli F, Garrido F. Induction of a different immune response in non-titanized compared to titanized polypropylene meshes. Acta Biomater 2023; 169:363-371. [PMID: 37579913 DOI: 10.1016/j.actbio.2023.08.018] [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: 02/27/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023]
Abstract
It is well known that pelvic organ prolapse (POP) significantly reduces the quality of life of affected women and in many cases requires corrective surgery. Aim of the study was to compare the immune response against titanized versus non-titanized meshes, especially macrophage polarization and immune checkpoint association. For this, we analyzed 644 POP surgeries, which were performed between 2017 and 2022, in our department. Four of them needed revision surgery caused by erosion. We analyzed the influx of CD68 & CD163 positive macrophages and the expression of immune checkpoint molecules PD-L1 and PD1 in these 4 patients. We identified a large number of CD68 and CD163 positive macrophages and additionally a PD-L1 expression of these cells. Based on the in-vivo results, we isolated monocytes and co-cultivated monocytes with different mesh material covered with or without fibroblasts. We identified a significantly enhanced macrophage activation and PD-L1 expression in macrophages surrounding non-titanized polypropylene mesh material. Encapsulation of the material by fibroblasts was crucial for that. Specifically, CD68-positive macrophages are upregulated (p < 0.001), co-expressing PD-L1 (p < 0.001) in monocytes co-cultivated with non-titanized polypropylene meshes. Monocytes co-cultivated with titanized polypropylene meshes showed significantly lower expression of CD163 (p = 0.027) and PD-L1 (p = 0.022). In conclusion, our in vitro data suggest that the titanium coating leads to a decreased polarization of macrophages and to a decreased immune response compared to non-titanized meshes. This could be an indication for the increased incidence of erosion of the non-titanized meshes, which is a severe complication of this procedure and requires revision surgery. STATEMENT OF SIGNIFICANCE: Pelvic organ prolapse is a well-known problem for women and often requires corrective surgery. Polypropylene meshes are often used, which differ in their coating (titanized vs. non-titanized). A severe side effect of these surgeries is mesh erosion, due to onset of inflammation, which requires revision surgery. We examined all erosion cases (4 of 644 patients) with implanted nontitanium-coated meshes by immunohistochemistry and found upregulation of macrophage polarization (as markers CD68 and CD163) and increased expression of the immune checkpoint molecules PD-L1 and PD1. This suggests inflammatory processes and an enhanced immune response. In addition, we set up an in vitro experiment to investigate whether coating plays a role. Here, we demonstrated that the non-titanized meshes elicited a significantly higher immune response in comparison to titanized meshes, which could lead to the higher erosion rate of the non-titanized meshes. Our results highlight the benefit of titanized meshes, which should lead to a lower revision surgery rate and thus improved patient outcome.
Collapse
Affiliation(s)
- Oleksii Protsepko
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, Augsburg 86156, Germany
| | - Philipp Voisard
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, Augsburg 86156, Germany
| | - Christina Kuhn
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, Augsburg 86156, Germany
| | - Andrea Maccagno
- Department of Pathology, University Hospital Augsburg, Stenglinstrasse 2, Augsburg 86156, Germany
| | - Christian Dannecker
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, Augsburg 86156, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, Augsburg 86156, Germany.
| | - Friedrich Pauli
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, Augsburg 86156, Germany
| | - Fabian Garrido
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, Augsburg 86156, Germany
| |
Collapse
|
40
|
Dutta SD, Ganguly K, Patil TV, Randhawa A, Lim KT. Unraveling the potential of 3D bioprinted immunomodulatory materials for regulating macrophage polarization: State-of-the-art in bone and associated tissue regeneration. Bioact Mater 2023; 28:284-310. [PMID: 37303852 PMCID: PMC10248805 DOI: 10.1016/j.bioactmat.2023.05.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/29/2023] [Accepted: 05/20/2023] [Indexed: 06/13/2023] Open
Abstract
Macrophage-assisted immunomodulation is an alternative strategy in tissue engineering, wherein the interplay between pro-inflammatory and anti-inflammatory macrophage cells and body cells determines the fate of healing or inflammation. Although several reports have demonstrated that tissue regeneration depends on spatial and temporal regulation of the biophysical or biochemical microenvironment of the biomaterial, the underlying molecular mechanism behind immunomodulation is still under consideration for developing immunomodulatory scaffolds. Currently, most fabricated immunomodulatory platforms reported in the literature show regenerative capabilities of a particular tissue, for example, endogenous tissue (e.g., bone, muscle, heart, kidney, and lungs) or exogenous tissue (e.g., skin and eye). In this review, we briefly introduced the necessity of the 3D immunomodulatory scaffolds and nanomaterials, focusing on material properties and their interaction with macrophages for general readers. This review also provides a comprehensive summary of macrophage origin and taxonomy, their diverse functions, and various signal transduction pathways during biomaterial-macrophage interaction, which is particularly helpful for material scientists and clinicians for developing next-generation immunomodulatory scaffolds. From a clinical standpoint, we briefly discussed the role of 3D biomaterial scaffolds and/or nanomaterial composites for macrophage-assisted tissue engineering with a special focus on bone and associated tissues. Finally, a summary with expert opinion is presented to address the challenges and future necessity of 3D bioprinted immunomodulatory materials for tissue engineering.
Collapse
Affiliation(s)
- Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V. Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
41
|
Falcone N, Ermis M, Tamay DG, Mecwan M, Monirizad M, Mathes TG, Jucaud V, Choroomi A, de Barros NR, Zhu Y, Vrana NE, Kraatz HB, Kim HJ, Khademhosseini A. Peptide Hydrogels as Immunomaterials and Their Use in Cancer Immunotherapy Delivery. Adv Healthc Mater 2023; 12:e2301096. [PMID: 37256647 PMCID: PMC10615713 DOI: 10.1002/adhm.202301096] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/15/2023] [Indexed: 06/01/2023]
Abstract
Peptide-based hydrogel biomaterials have emerged as an excellent strategy for immune system modulation. Peptide-based hydrogels are supramolecular materials that self-assemble into various nanostructures through various interactive forces (i.e., hydrogen bonding and hydrophobic interactions) and respond to microenvironmental stimuli (i.e., pH, temperature). While they have been reported in numerous biomedical applications, they have recently been deemed promising candidates to improve the efficacy of cancer immunotherapies and treatments. Immunotherapies seek to harness the body's immune system to preemptively protect against and treat various diseases, such as cancer. However, their low efficacy rates result in limited patient responses to treatment. Here, the immunomaterial's potential to improve these efficacy rates by either functioning as immune stimulators through direct immune system interactions and/or delivering a range of immune agents is highlighted. The chemical and physical properties of these peptide-based materials that lead to immuno modulation and how one may design a system to achieve desired immune responses in a controllable manner are discussed. Works in the literature that reports peptide hydrogels as adjuvant systems and for the delivery of immunotherapies are highlighted. Finally, the future trends and possible developments based on peptide hydrogels for cancer immunotherapy applications are discussed.
Collapse
Affiliation(s)
- Natashya Falcone
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, 06800, Turkey
| | - Dilara Goksu Tamay
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, 06800, Turkey
- Department of Biotechnology, Middle East Technical University, Ankara, 06800, Turkey
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Mahsa Monirizad
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Tess Grett Mathes
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Auveen Choroomi
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Nihal Engin Vrana
- SPARTHA Medical, CRBS 1 Rue Eugene Boeckel, Strasbourg, 67000, France
| | - Heinz-Bernhard Kraatz
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, M5S 3E5, Canada
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, ON, M1C 1A4, Canada
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| |
Collapse
|
42
|
Alvarez-Lorenzo C, Zarur M, Seijo-Rabina A, Blanco-Fernandez B, Rodríguez-Moldes I, Concheiro A. Physical stimuli-emitting scaffolds: The role of piezoelectricity in tissue regeneration. Mater Today Bio 2023; 22:100740. [PMID: 37521523 PMCID: PMC10374602 DOI: 10.1016/j.mtbio.2023.100740] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/01/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023] Open
Abstract
The imbalance between life expectancy and quality of life is increasing due to the raising prevalence of chronic diseases. Musculoskeletal disorders and chronic wounds affect a growing percentage of people and demand more efficient tools for regenerative medicine. Scaffolds that can better mimic the natural physical stimuli that tissues receive under healthy conditions and during healing may significantly aid the regeneration process. Shape, mechanical properties, pore size and interconnectivity have already been demonstrated to be relevant scaffold features that can determine cell adhesion and differentiation. Much less attention has been paid to scaffolds that can deliver more dynamic physical stimuli, such as electrical signals. Recent developments in the precise measurement of electrical fields in vivo have revealed their key role in cell movement (galvanotaxis), growth, activation of secondary cascades, and differentiation to different lineages in a variety of tissues, not just neural. Piezoelectric scaffolds can mimic the natural bioelectric potentials and gradients in an autonomous way by generating the electric stimuli themselves when subjected to mechanical loads or, if the patient or the tissue lacks mobility, ultrasound irradiation. This review provides an analysis on endogenous bioelectrical signals, recent developments on piezoelectric scaffolds for bone, cartilage, tendon and nerve regeneration, and their main outcomes in vivo. Wound healing with piezoelectric dressings is addressed in the last section with relevant examples of performance in animal models. Results evidence that a fine adjustment of material composition and processing (electrospinning, corona poling, 3D printing, annealing) provides scaffolds that act as true emitters of electrical stimuli that activate endogenous signaling pathways for more efficient and long-term tissue repair.
Collapse
Affiliation(s)
- Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Mariana Zarur
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Alejandro Seijo-Rabina
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Barbara Blanco-Fernandez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Isabel Rodríguez-Moldes
- Grupo NEURODEVO, Departamento de Bioloxía Funcional, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| |
Collapse
|
43
|
Unzai T, Washisaka T, Tabata Y. An artificial silk elastin-like protein modifies the polarization of human macrophages line THP-1. J Biomater Appl 2023; 38:361-371. [PMID: 37494553 DOI: 10.1177/08853282231192186] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
A silk elastin-like protein (SELP) is an artificial compound with silk fibroin-like and elastin-like tandem repeats. The objective of this study is to evaluate the influence of SELP on the polarization of human monocytoma cell line (THP-1)-derived macrophages. When the macrophages of inflammation-type (M1) were cultured with different concentrations of SELP solution, the secretion of a pro-inflammatory cytokine, tumor necrotizing factor (TNF) -α was significantly suppressed at the higher concentrations. In addition, the secretion of an anti-inflammation cytokine, interleukin (IL)-10, was significantly enhanced from the macrophage of M0-, M1-, and M2-types. By the incubation with soluble SELP, the morphology of M2-type macrophages changed to be of an extended shape. Following incubation with the sponge of SELP, M0-type macrophages secreted IL-10 with time. It is concluded that the SELP itself in solution has an ability to induce the anti-inflammation of M2-type macrophages.
Collapse
Affiliation(s)
- Tomo Unzai
- Laboratory of Biomaterials, Kyoto University Institute for Life and Medical Sciences, Kyoto, Japan
| | - Taichi Washisaka
- Laboratory of Biomaterials, Kyoto University Institute for Life and Medical Sciences, Kyoto, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Kyoto University Institute for Life and Medical Sciences, Kyoto, Japan
| |
Collapse
|
44
|
Scheuermann K, Viana CTR, Dos Reis DC, de Lazari MGT, Orellano LAA, Machado CT, Dos Santos LCC, Ulrich H, Capettini LSA, Andrade SP, Campos PP. Amitriptyline efficacy in decreasing implant-induced foreign body reaction. IUBMB Life 2023; 75:732-742. [PMID: 37086464 DOI: 10.1002/iub.2725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 03/15/2023] [Indexed: 04/24/2023]
Abstract
Beyond its actions on the nervous system, amitriptyline (AM) has been shown to lower inflammatory, angiogenic, and fibrogenic markers in a few pathological conditions in human and in experimental animal models. However, its effects on foreign body reaction (FBR), a complex adverse healing process, after biomedical material implantation are not known. We have evaluated the effects of AM on the angiogenic and fibrogenic components on a model of implant-induced FBR. Sponge disks were implanted subcutaneously in C57BL/6 mice, that were treated daily with oral administration of AM (5 mg/kg) for seven consecutive days in two protocols: treatment was started on the day of surgery and the implants were removed on the seventh day after implantation and treatment started 7 days after implantation and the implants removed 14 after implantation. None of the angiogenic (vessels, Vascular endothelial growth factor (VEGF), and interleukin-1β (IL-1β) or fibrogenic parameters (collagen, TGF-β, and fibrous capsule) and giant cell numbers analyzed were attenuated by AM in 7-day-old implants. However, AM was able to downregulate angiogenesis and FBR in 14-day-old implants. The effects of AM described here expands its range of actions as a potential agent capable of attenuating fibroproliferative processes that may impair functionality of implantable devices.
Collapse
Affiliation(s)
- Karina Scheuermann
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Celso Tarso Rodrigues Viana
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Diego Carlos Dos Reis
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Laura Alejandra Ariza Orellano
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Clara Tolentino Machado
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, São Paulo, Brazil
| | | | - Silvia Passos Andrade
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Paula Peixoto Campos
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
45
|
Barbault F, Brémond E, Rey J, Tufféry P, Maurel F. DockSurf: A Molecular Modeling Software for the Prediction of Protein/Surface Adhesion. J Chem Inf Model 2023; 63:5220-5231. [PMID: 37579187 DOI: 10.1021/acs.jcim.3c00569] [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: 08/16/2023]
Abstract
The elucidation of structural interfaces between proteins and inorganic surfaces is a crucial aspect of bionanotechnology development. Despite its significance, the interfacial structures between proteins and metallic surfaces are yet to be fully understood, and the lack of experimental investigation has impeded the development of many devices. To overcome this limitation, we suggest considering the generation of protein/surface structures as a molecular docking problem with a homogenous plan as the target. To this extent, we propose a new software, DockSurf, which aims to quickly propose reliable protein/surface structures. Our approach considers the conformational exploration with Euler's angles, which provide a cartography instead of a unique structure. Interaction energies were derived from quantum mechanics computations for a set of small molecules that describe protein atom types and implemented in a Derjaguin, Landau, Verwey, and Overbeek potential for the consideration of large systems such as proteins. The validation of DockSurf software was conducted with molecular dynamics for corona proteins with gold surfaces and provided enthusiastic results. This software is implemented in the RPBS platform to facilitate widespread access to the scientific community.
Collapse
Affiliation(s)
| | - Eric Brémond
- Université Paris Cité, CNRS, ITODYS, F-75013 Paris, France
| | - Julien Rey
- Université Paris Cité, CNRS UMR 8251, INSERM U1133, RPBS, 75205 Paris, France
| | - Pierre Tufféry
- Université Paris Cité, CNRS UMR 8251, INSERM U1133, RPBS, 75205 Paris, France
| | | |
Collapse
|
46
|
Zhao Z, Zhao Q, Chen H, Chen F, Wang F, Tang H, Xia H, Zhou Y, Sun Y. Role of dendritic cells in MYD88-mediated immune recognition and osteoinduction initiated by the implantation of biomaterials. Int J Oral Sci 2023; 15:31. [PMID: 37532700 PMCID: PMC10397189 DOI: 10.1038/s41368-023-00234-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/21/2023] [Accepted: 06/26/2023] [Indexed: 08/04/2023] Open
Abstract
Bone substitute material implantation has become an important treatment strategy for the repair of oral and maxillofacial bone defects. Recent studies have shown that appropriate inflammatory and immune cells are essential factors in the process of osteoinduction of bone substitute materials. Previous studies have mainly focused on innate immune cells such as macrophages. In our previous work, we found that T lymphocytes, as adaptive immune cells, are also essential in the osteoinduction procedure. As the most important antigen-presenting cell, whether dendritic cells (DCs) can recognize non-antigen biomaterials and participate in osteoinduction was still unclear. In this study, we found that surgical trauma associated with materials implantation induces necrocytosis, and this causes the release of high mobility group protein-1 (HMGB1), which is adsorbed on the surface of bone substitute materials. Subsequently, HMGB1-adsorbed materials were recognized by the TLR4-MYD88-NFκB signal axis of dendritic cells, and the inflammatory response was activated. Finally, activated DCs release regeneration-related chemokines, recruit mesenchymal stem cells, and initiate the osteoinduction process. This study sheds light on the immune-regeneration process after bone substitute materials implantation, points out a potential direction for the development of bone substitute materials, and provides guidance for the development of clinical surgical methods.
Collapse
Affiliation(s)
- Zifan Zhao
- Center of Digital Dentistry, Faculty of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Qin Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hu Chen
- Center of Digital Dentistry, Faculty of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Fanfan Chen
- Center of Digital Dentistry, Faculty of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Feifei Wang
- Center of Digital Dentistry, Faculty of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Hua Tang
- Institute of Infection and Immunity, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Haibin Xia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yongsheng Zhou
- Center of Digital Dentistry, Faculty of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China.
| | - Yuchun Sun
- Center of Digital Dentistry, Faculty of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China.
| |
Collapse
|
47
|
Ben Amara H, Martinez DC, Shah FA, Loo AJ, Emanuelsson L, Norlindh B, Willumeit-Römer R, Plocinski T, Swieszkowski W, Palmquist A, Omar O, Thomsen P. Magnesium implant degradation provides immunomodulatory and proangiogenic effects and attenuates peri-implant fibrosis in soft tissues. Bioact Mater 2023; 26:353-369. [PMID: 36942009 PMCID: PMC10024189 DOI: 10.1016/j.bioactmat.2023.02.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 03/17/2023] Open
Abstract
Implants made of magnesium (Mg) are increasingly employed in patients to achieve osteosynthesis while degrading in situ. Since Mg implants and Mg2+ have been suggested to possess anti-inflammatory properties, the clinically observed soft tissue inflammation around Mg implants is enigmatic. Here, using a rat soft tissue model and a 1-28 d observation period, we determined the temporo-spatial cell distribution and behavior in relation to sequential changes of pure Mg implant surface properties and Mg2+ release. Compared to nondegradable titanium (Ti) implants, Mg degradation exacerbated initial inflammation. Release of Mg degradation products at the tissue-implant interface, culminating at 3 d, actively initiated chemotaxis and upregulated mRNA and protein immunomodulatory markers, particularly inducible nitric oxide synthase and toll-like receptor-4 up to 6 d, yet without a cytotoxic effect. Increased vascularization was demonstrated morphologically, preceded by high expression of vascular endothelial growth factor. The transition to appropriate tissue repair coincided with implant surface enrichment of Ca and P and reduced peri-implant Mg2+ concentration. Mg implants revealed a thinner fibrous encapsulation compared with Ti. The detailed understanding of the relationship between Mg material properties and the spatial and time-resolved cellular processes provides a basis for the interpretation of clinical observations and future tailoring of Mg implants.
Collapse
Affiliation(s)
- Heithem Ben Amara
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Diana C. Martinez
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Furqan A. Shah
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Johansson Loo
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lena Emanuelsson
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Birgitta Norlindh
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Tomasz Plocinski
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Wojciech Swieszkowski
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Anders Palmquist
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Omar Omar
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter Thomsen
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Corresponding author. Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg Box 412, SE-405 30, Gothenburg, Sweden.
| |
Collapse
|
48
|
Han F, Meng Q, Xie E, Li K, Hu J, Chen Q, Li J, Han F. Engineered biomimetic micro/nano-materials for tissue regeneration. Front Bioeng Biotechnol 2023; 11:1205792. [PMID: 37469449 PMCID: PMC10352664 DOI: 10.3389/fbioe.2023.1205792] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/26/2023] [Indexed: 07/21/2023] Open
Abstract
The incidence of tissue and organ damage caused by various diseases is increasing worldwide. Tissue engineering is a promising strategy of tackling this problem because of its potential to regenerate or replace damaged tissues and organs. The biochemical and biophysical cues of biomaterials can stimulate and induce biological activities such as cell adhesion, proliferation and differentiation, and ultimately achieve tissue repair and regeneration. Micro/nano materials are a special type of biomaterial that can mimic the microstructure of tissues on a microscopic scale due to its precise construction, further providing scaffolds with specific three-dimensional structures to guide the activities of cells. The study and application of biomimetic micro/nano-materials have greatly promoted the development of tissue engineering. This review aims to provide an overview of the different types of micro/nanomaterials, their preparation methods and their application in tissue regeneration.
Collapse
Affiliation(s)
- Feng Han
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Qingchen Meng
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - En Xie
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Kexin Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Jie Hu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Qianglong Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Jiaying Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Fengxuan Han
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang, China
| |
Collapse
|
49
|
Zhang P, Liu Y, Feng G, Li C, Zhou J, Du C, Bai Y, Hu S, Huang T, Wang G, Quan P, Hirvonen J, Fan J, Santos HA, Liu D. Controlled Interfacial Polymer Self-Assembly Coordinates Ultrahigh Drug Loading and Zero-Order Release in Particles Prepared under Continuous Flow. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211254. [PMID: 36802103 DOI: 10.1002/adma.202211254] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/11/2023] [Indexed: 06/02/2023]
Abstract
Microparticles are successfully engineered through controlled interfacial self-assembly of polymers to harmonize ultrahigh drug loading with zero-order release of protein payloads. To address their poor miscibility with carrier materials, protein molecules are transformed into nanoparticles, whose surfaces are covered with polymer molecules. This polymer layer hinders the transfer of cargo nanoparticles from oil to water, achieving superior encapsulation efficiency (up to 99.9%). To control payload release, the polymer density at the oil-water interface is enhanced, forming a compact shell for microparticles. The resultant microparticles can harvest up to 49.9% mass fraction of proteins with zero-order release kinetics in vivo, enabling an efficient glycemic control in type 1 diabetes. Moreover, the precise control of engineering process offered through continuous flow results in high batch-to-batch reproducibility and, ultimately, excellent scale-up feasibility.
Collapse
Affiliation(s)
- Pei Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| | - Yingxin Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Guobing Feng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Cong Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jun Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Chunyang Du
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuancheng Bai
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Shuai Hu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Tianhe Huang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Guan Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Peng Quan
- Department of Pharmaceutical Science, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| | - Jin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Dongfei Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China
| |
Collapse
|
50
|
Miron A, Giurcaneanu C, Mihai MM, Beiu C, Voiculescu VM, Popescu MN, Soare E, Popa LG. Antimicrobial Biomaterials for Chronic Wound Care. Pharmaceutics 2023; 15:1606. [PMID: 37376055 DOI: 10.3390/pharmaceutics15061606] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/09/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic wounds encompass a myriad of lesions, including venous and arterial leg ulcers, diabetic foot ulcers (DFUs), pressure ulcers, non-healing surgical wounds and others. Despite the etiological differences, chronic wounds share several features at a molecular level. The wound bed is a convenient environment for microbial adherence, colonization and infection, with the initiation of a complex host-microbiome interplay. Chronic wound infections with mono- or poly-microbial biofilms are frequent and their management is challenging due to tolerance and resistance to antimicrobial therapy (systemic antibiotic or antifungal therapy or antiseptic topicals) and to the host's immune defense mechanisms. The ideal dressing should maintain moisture, allow water and gas permeability, absorb wound exudates, protect against bacteria and other infectious agents, be biocompatible, be non-allergenic, be non-toxic and biodegradable, be easy to use and remove and, last but not least, it should be cost-efficient. Although many wound dressings possess intrinsic antimicrobial properties acting as a barrier to pathogen invasion, adding anti-infectious targeted agents to the wound dressing may increase their efficiency. Antimicrobial biomaterials may represent a potential substitute for systemic treatment of chronic wound infections. In this review, we aim to describe the available types of antimicrobial biomaterials for chronic wound care and discuss the host response and the spectrum of pathophysiologic changes resulting from the contact between biomaterials and host tissues.
Collapse
Affiliation(s)
- Adrian Miron
- Department of General Surgery, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, No. 37 Dionisie Lupu Str., 030167 Bucharest, Romania
- Clinic of General Surgery, Elias Emergency University Hospital, No. 17 Marasti Blvd., 011461 Bucharest, Romania
| | - Calin Giurcaneanu
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, No. 37 Dionisie Lupu Str., 030167 Bucharest, Romania
- Clinic of Dermatology, Elias Emergency University Hospital, No. 17 Marasti Blvd., 011461 Bucharest, Romania
| | - Mara Madalina Mihai
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, No. 37 Dionisie Lupu Str., 030167 Bucharest, Romania
- Clinic of Dermatology, Elias Emergency University Hospital, No. 17 Marasti Blvd., 011461 Bucharest, Romania
- Department of Microbiology, Faculty of Biology, ICUB-Research Institute, University of Bucharest, No. 90 Panduri Str., 050663 Bucharest, Romania
| | - Cristina Beiu
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, No. 37 Dionisie Lupu Str., 030167 Bucharest, Romania
- Clinic of Dermatology, Elias Emergency University Hospital, No. 17 Marasti Blvd., 011461 Bucharest, Romania
| | - Vlad Mihai Voiculescu
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, No. 37 Dionisie Lupu Str., 030167 Bucharest, Romania
- Clinic of Dermatology, Elias Emergency University Hospital, No. 17 Marasti Blvd., 011461 Bucharest, Romania
| | - Marius Nicolae Popescu
- Department of Microbiology, Faculty of Biology, ICUB-Research Institute, University of Bucharest, No. 90 Panduri Str., 050663 Bucharest, Romania
- Department of Physical and Rehabilitation Medicine, Carol Davila University of Medicine and Pharmacy, No. 37 Dionisie Lupu Str., 030167 Bucharest, Romania
- Clinic of Physical and Rehabilitation Medicine, Elias Emergency University Hospital, No. 17 Marasti Blvd., 011461 Bucharest, Romania
| | - Elena Soare
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, No. 37 Dionisie Lupu Str., 030167 Bucharest, Romania
| | - Liliana Gabriela Popa
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, No. 37 Dionisie Lupu Str., 030167 Bucharest, Romania
- Clinic of Dermatology, Elias Emergency University Hospital, No. 17 Marasti Blvd., 011461 Bucharest, Romania
| |
Collapse
|