1
|
Pan X, Zong Q, Liu C, Wu H, Fu B, Wang Y, Sun W, Zhai Y. Konjac glucomannan exerts regulatory effects on macrophages and its applications in biomedical engineering. Carbohydr Polym 2024; 345:122571. [PMID: 39227106 DOI: 10.1016/j.carbpol.2024.122571] [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: 06/27/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024]
Abstract
Konjac glucomannan (KGM) molecular chains contain a small amount of acetyl groups and a large number of hydroxyl groups, thereby exhibiting exceptional water retention and gel-forming properties. To meet diverse requirements, KGM undergoes modification processes such as oxidation, acetylation, grafting, and cationization, which reduce its viscosity, enhance its mechanical strength, and improve its water solubility. Researchers have found that KGM and its derivatives can regulate the polarization of macrophages, inducing their transformation into classically activated M1-type macrophages or alternatively activated M2-type macrophages, and even facilitating the interconversion between M1 and M2 phenotypes. Concurrently, the modulation of macrophage polarization states holds significant importance for chronic wound healing, inflammatory bowel disease (IBD), antitumor therapy, tissue engineering scaffolds, oral vaccines, pulmonary delivery, and probiotics. Therefore, KGM has the advantages of both immunomodulatory effects (biological activity) and gel-forming properties (physicochemical properties), giving it significant advantages in a variety of biomedical engineering applications.
Collapse
Affiliation(s)
- Xi Pan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qida Zong
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chun Liu
- Hainan Institute for Drug Control, Haikou 570311, China
| | - Huiying Wu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bo Fu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ye Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wei Sun
- Department of Biomedical Engineering, School of Pharmaceutical University, Shenyang 110016, China.
| | - Yinglei Zhai
- Department of Biomedical Engineering, School of Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
2
|
Tang Y, Zhou Y, Zhang M. A Chitosan Scaffold Supports the Enhanced and Prolonged Differentiation of HiPSCs into Nucleus Pulposus-like Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28263-28275. [PMID: 38788694 DOI: 10.1021/acsami.4c06013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Intervertebral disc degeneration (IDD) is a progressive condition and stands as one of the primary causes of low back pain. Cell therapy that uses nucleus pulposus (NP)-like cells derived from human induced pluripotent stem cells (hiPSCs) holds great promise as a treatment for IDD. However, the conventional two-dimensional (2D) monolayer cultures oversimplify cell-cell interactions, leading to suboptimal differentiation efficiency and potential loss of phenotype. While three-dimensional (3D) culture systems like Matrigel improve hiPSC differentiation efficiency, they are limited by animal-derived materials for translation, poorly defined composition, short-term degradation, and high cost. In this study, we introduce a new 3D scaffold fabricated using medical-grade chitosan with a high degree of deacetylation. The scaffold features a highly interconnected porous structure, near-neutral surface charge, and exceptional degradation stability, benefiting iPSC adhesion and proliferation. This scaffold remarkably enhances the differentiation efficiency and allows uninterrupted differentiation for up to 25 days without subculturing. Notably, cells differentiated on the chitosan scaffold exhibited increased cell survival rates and upregulated gene expression associated with extracellular matrix secretion under a chemically defined condition mimicking the challenging microenvironment of intervertebral discs. These characteristics qualify the chitosan scaffold-cell construct for direct implantation, serving as both a structural support and a cellular source for enhanced stem cell therapy for IDD.
Collapse
Affiliation(s)
- Yuanzhang Tang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Yang Zhou
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
3
|
Chen Y, Luo Z, Meng W, Liu K, Chen Q, Cai Y, Ding Z, Huang C, Zhou Z, Jiang M, Zhou L. Decoding the "Fingerprint" of Implant Materials: Insights into the Foreign Body Reaction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310325. [PMID: 38191783 DOI: 10.1002/smll.202310325] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/12/2023] [Indexed: 01/10/2024]
Abstract
Foreign body reaction (FBR) is a prevalent yet often overlooked pathological phenomenon, particularly within the field of biomedical implantation. The presence of FBR poses a heavy burden on both the medical and socioeconomic systems. This review seeks to elucidate the protein "fingerprint" of implant materials, which is generated by the physiochemical properties of the implant materials themselves. In this review, the activity of macrophages, the formation of foreign body giant cells (FBGCs), and the development of fibrosis capsules in the context of FBR are introduced. Additionally, the relationship between various implant materials and FBR is elucidated in detail, as is an overview of the existing approaches and technologies employed to alleviate FBR. Finally, the significance of implant components (metallic materials and non-metallic materials), surface CHEMISTRY (charge and wettability), and physical characteristics (topography, roughness, and stiffness) in establishing the protein "fingerprint" of implant materials is also well documented. In conclusion, this review aims to emphasize the importance of FBR on implant materials and provides the current perspectives and approaches in developing implant materials with anti-FBR properties.
Collapse
Affiliation(s)
- Yangmengfan Chen
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zeyu Luo
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Weikun Meng
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kai Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiqing Chen
- Department of Ultrasound, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| | - Yongrui Cai
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zichuan Ding
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chao Huang
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zongke Zhou
- Orthopedic Research Institution, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Jiang
- Emergency and Trauma Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Liqiang Zhou
- MOE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| |
Collapse
|
4
|
Schluck M, Weiden J, Verdoes M, Figdor CG. Insights in the host response towards biomaterial-based scaffolds for cancer therapy. Front Bioeng Biotechnol 2023; 11:1149943. [PMID: 37342507 PMCID: PMC10277494 DOI: 10.3389/fbioe.2023.1149943] [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: 01/23/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Immunotherapeutic strategies have shown promising results in the treatment of cancer. However, not all patients respond, and treatments can have severe side-effects. Adoptive cell therapy (ACT) has shown remarkable therapeutic efficacy across different leukaemia and lymphoma types. But the treatment of solid tumours remains a challenge due to limited persistence and tumour infiltration. We believe that biomaterial-based scaffolds are promising new tools and may address several of the challenges associated with cancer vaccination and ACT. In particular, biomaterial-based scaffold implants allow for controlled delivery of activating signals and/or functional T cells at specific sites. One of the main challenges for their application forms the host response against these scaffolds, which includes unwanted myeloid cell infiltration and the formation of a fibrotic capsule around the scaffold, thereby limiting cell traffic. In this review we provide an overview of several of the biomaterial-based scaffolds designed for cancer therapy to date. We will discuss the host responses observed and we will highlight design parameters that influence this response and their potential impact on therapeutic outcome.
Collapse
Affiliation(s)
- Marjolein Schluck
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Oncode Institute, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Jorieke Weiden
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Oncode Institute, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Carl G. Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Oncode Institute, Nijmegen, Netherlands
- Institute for Chemical Immunology, Nijmegen, Netherlands
| |
Collapse
|
5
|
Hansda A, Mukherjee S, Dixit K, Dhara S, Mukherjee G. Immunological Perspectives Involved in Tissue Engineering. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
6
|
Rahyussalim AJ, Aprilya D, Handidwiono R, Whulanza Y, Ramahdita G, Kurniawati T. The Use of 3D Polylactic Acid Scaffolds with Hydroxyapatite/Alginate Composite Injection and Mesenchymal Stem Cells as Laminoplasty Spacers in Rabbits. Polymers (Basel) 2022; 14:polym14163292. [PMID: 36015548 PMCID: PMC9416571 DOI: 10.3390/polym14163292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/29/2022] [Accepted: 08/07/2022] [Indexed: 11/16/2022] Open
Abstract
Several types of laminoplasty spacer have been used to fill bone gaps and maintain a widened canal. A 3D scaffold can be used as an alternative spacer to minimize the risk observed in allografts or autografts. This study aims to evaluate the in vivo biocompatibility and tissue−scaffold integration of a polylactic acid (PLA) scaffold with the addition of alginate/hydroxyapatite (HA) and mesenchymal stem cell (MSc) injections. This is an experimental study with a pretest and post-test control group design. A total of 15 laminoplasty rabbit models were divided into five groups with variations in the autograft, PLA, HA/alginate, and MSc scaffold. In general, there were no signs of inflammation in most samples (47%), and there were no samples with areas of necrosis. There were no significant differences in the histopathological results and microstructural assessment between the five groups. This demonstrates that the synthetic scaffolds that we used had a similar tissue reaction and tissue integration profile as the autograft (p > 0.05). We recommend further translational studies in humans so that this biocompatible fabricated scaffold can be used to fill bone defects.
Collapse
Affiliation(s)
- Ahmad Jabir Rahyussalim
- Department of Orthopaedic & Traumatology, Cipto Mangunkusumo National General Hospital and Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo General Hospital, Jakarta 10430, Indonesia
- Stem Cells and Tissue Engineering Research Cluster, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Dina Aprilya
- Department of Orthopaedic & Traumatology, Cipto Mangunkusumo National General Hospital and Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Correspondence:
| | - Raden Handidwiono
- Department of Orthopaedic & Traumatology, Cipto Mangunkusumo National General Hospital and Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Yudan Whulanza
- Department of Mechanical Engineering, Faculty of Engineering, Universitas Indonesia, Depok 16424, Indonesia
- Research Center for Biomedical Engineering, Faculty of Engineering, Universitas Indonesia, Depok 16424, Indonesia
| | - Ghiska Ramahdita
- Mechanical Engineering and Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Tri Kurniawati
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo General Hospital, Jakarta 10430, Indonesia
- Stem Cells and Tissue Engineering Research Cluster, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| |
Collapse
|
7
|
You J, Zhang Y, Zhou Y. Strontium Functionalized in Biomaterials for Bone Tissue Engineering: A Prominent Role in Osteoimmunomodulation. Front Bioeng Biotechnol 2022; 10:928799. [PMID: 35875505 PMCID: PMC9298737 DOI: 10.3389/fbioe.2022.928799] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/13/2022] [Indexed: 12/24/2022] Open
Abstract
With the development of bone tissue engineering bio-scaffold materials by adding metallic ions to improve bone healing have been extensively explored in the past decades. Strontium a non-radioactive element, as an essential osteophilic trace element for the human body, has received widespread attention in the medical field due to its superior biological properties of inhibiting bone resorption and promoting osteogenesis. As the concept of osteoimmunology developed, the design of orthopedic biomaterials has gradually shifted from “immune-friendly” to “immunomodulatory” with the aim of promoting bone healing by modulating the immune microenvironment through implanted biomaterials. The process of bone healing can be regarded as an immune-induced procedure in which immune cells can target the effector cells such as macrophages, neutrophils, osteocytes, and osteoprogenitor cells through paracrine mechanisms, affecting pathological alveolar bone resorption and physiological bone regeneration. As a kind of crucial immune cell, macrophages play a critical role in the early period of wound repair and host defense after biomaterial implantation. Despite Sr-doped biomaterials being increasingly investigated, how extracellular Sr2+ guides the organism toward favorable osteogenesis by modulating macrophages in the bone tissue microenvironment has rarely been studied. This review focuses on recent knowledge that the trace element Sr regulates bone regeneration mechanisms through the regulation of macrophage polarization, which is significant for the future development of Sr-doped bone repair materials. We will also summarize the primary mechanism of Sr2+ in bone, including calcium-sensing receptor (CaSR) and osteogenesis-related signaling pathways.
Collapse
Affiliation(s)
- Jiaqian You
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yidi Zhang
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yanmin Zhou
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
8
|
Bai H, Wang Y, Zhao Y, Chen X, Xiao Y, Bao C. HIF signaling: A new propellant in bone regeneration. BIOMATERIALS ADVANCES 2022; 138:212874. [PMID: 35913258 DOI: 10.1016/j.bioadv.2022.212874] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/04/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
Bone tissue destruction leads to severe pain, physical flaws, and loss of motility. Bone repair using biocompatible and osteo-inductive scaffolds is regarded as a viable and potential therapeutic approach. However, for large-scale bone regeneration, oxygen and nutrient supply have become limiting factors. Further, a considerable need exists for recruited cell activities and blood vessel growth. Hypoxia-inducible factor (HIF) signaling pathways induced by hypoxia are involved in angiogenesis and osteogenesis. As an important transcription factor, HIF-1 functions by modulating vital genes, such as VEGF, PDK1, and EPO, and is a crucial regulator that influences the final fate of bone regeneration. Collectively, to achieve better osteogenesis results, the in-depth molecular mechanisms that underpin the links between materials, cells, and HIF signaling pathways must be determined. This review aimed to provide an in-depth insight into recent progress in HIF-regulated bone regeneration. Hypoxia and cellular oxygen-sensing mechanisms and their correlations with osteogenesis were determined, and recent studies on hypoxia-inducing and hypoxia-mimicking strategies were briefly described. Finally, the potential applications of HIF signaling in bone regeneration were highlighted. This review provides theoretical support for establishing a novel and viable bone repair strategy in the clinic by harnessing HIF signaling.
Collapse
Affiliation(s)
- Hetian Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China
| | - Yue Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China
| | - Yi Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China
| | - Xin Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China
| | - Yu Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China.
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, No. 14, Section 3, Renmin Nan Road, Chengdu 610041, Sichuan, China
| |
Collapse
|
9
|
Henckes NAC, Chuang L, Bosak I, Carazzai R, Garcez T, Kuhl CP, de Oliveira FDS, Loureiro Dos Santos LA, Visioli F, Cirne-Lima EO. Tissue engineering application combining epoxidized natural rubber blend and mesenchymal stem cells in in vivo response. J Biomater Appl 2022; 37:698-711. [PMID: 35733325 DOI: 10.1177/08853282221110476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study aimed to investigate biocompatibility, integration, and tissue host response of the Poly (Lactic-co-Glycolic acid) (PLGA)/Poly (isoprene) (PI) epoxidized (PLGA/PIepox) innovative scaffold combined with adipose derived mesenchymal stem cells (ADSC). We implanted the scaffold subcutaneously on the back of 18 female rats and monitored them for up to 14 days. When compared to controls, PLGA/PIepox + ADSC demonstrated an earlier vascularization, a tendency of inflammation reduction, an adequate tissue integration, higher cell proliferation, and a tendency of expression of collagen decreasing. However, 14 days post-implantation we found similar levels of CD31, Ki67 and AE1/AE3 in PLGA/PIepox when compared to control groups. The interesting results, lead us to the assumption that PLGA/PIepox is able to provide an effective delivery system for ADSC on tissue host. This animal study assesses PLGA/PIepox + ADSC in in vivo tissue functionality and validation of use, serving as a proof of concept for future clinical translation as it presents an innovative and promising tissue engineering opportunity for the use in tissue reconstruction.
Collapse
Affiliation(s)
- Nicole Andréa Corbellini Henckes
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-graduação em Ciências da Saúde: Ginecologia e Obstetrícia, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Laura Chuang
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Isadora Bosak
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Rafael Carazzai
- Laboratório de Biomateriais e Cerâmicas Avançadas, Departamento de Materiais, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tuane Garcez
- Unidade de Experimentação Animal - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Cristiana Palma Kuhl
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-graduação em Ciências da Saúde: Ginecologia e Obstetrícia, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Dos Santos de Oliveira
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Luis Alberto Loureiro Dos Santos
- Laboratório de Biomateriais e Cerâmicas Avançadas, Departamento de Materiais, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Visioli
- Unidade de Patologia Experimental - Centro de Pesquisa Experimental, 37895Hospital de Clinicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-graduação em Odontologia, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Elizabeth Obino Cirne-Lima
- Laboratório de Embriologia e Diferenciação Celular - Centro de Pesquisa Experimental, 37895Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Programa de Pós-graduação em Ciências da Saúde: Ginecologia e Obstetrícia, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Patologia Clínica Veterinária, Faculdade de Veterinária, 28124Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
10
|
[Experimental study on the construction of telmisartan/collagen/polycaprolactone nerve conduit and its repair effect on rat sciatic nerve defect]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2022; 36:352-361. [PMID: 35293178 PMCID: PMC8923921 DOI: 10.7507/1002-1892.202108142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To construction the telmisartan/collagen/polycaprolactone (PCL) nerve conduit and assess its effect on repairing sciatic nerve defect in rats. METHODS The 60% collagen/hexafluoroisopropanol (HFIP) solution and 40% PCL/HFIP solution were prepared and mixed (collagen/PCL solution). Then the 0, 5, 10, and 20 mg of telmisartan were mixed with the 10 mL collagen/PCL solution, respectively. Telmisartan/collagen/PCL nerve conduits were fabricated via high voltage electrospinning technology. The structure of nerve conduit before and after crosslinking was observed by using scanning electron microscope (SEM). The drug release efficiency was detected by in vitro sustained release method. RAW264.7 cells were cultured with lipopolysaccharide to induce inflammation, and then co-cultured with nerve conduits loaded with different concentrations of telmisartan for 24 hours. The mRNA expressions of inducible nitric oxide synthase (iNOS) and Arginase 1 (Arg-1) were detected by using real-time fluorescence quantitative PCR. Forty adult Wistar rats were randomly divided into 4 groups ( n=10). After preparing 15-mm-long sciatic nerve defect, the defect was repaired by cross-linked nerve conduits loaded with 0, 5, 10, and 20 mg telmisartan in groups A, B, C, and D, respectively. After operation, the general condition of rats was observed after operation; the sciatic function index (SFI) was tested; the bridging between the nerve conduit and sciatic nerve, and the integrity of nerve conduit were observed; the tissue growth in nerve conduit and material degradation were observed by HE staining; the expressions of CD86 (M1 macrophage marker), CD206 (M2 macrophage marker), myelin basic protein (MBP), and myelin protein 0 (P0) in new tissues were also observed by immunohistochemical staining; the expressions of neurofilament 200 (NF-200) and S-100β in new tissues were assessed by immunofluorescence staining. RESULTS The general observation showed that the inner diameter of the nerve conduit was 1.8 mm and the outer diameter was 2.0 mm. After cross-linking by genipin, the nanofiber became thicker and denser. The drug release test showed that the telmisartan loaded nerve conduit could be released gradually. With the increase of telmisartan content in nerve conduit, the iNOS mRNA expression decreased and the Arg-1 mRNA expression increased; and the differences between 20 mg group and other groups were significant ( P<0.05). In vivo experiment showed that all animals in each group survived until the completion of the experiment. The SFI was significantly higher in groups C and D than in groups A and B at different time points ( P<0.05) and in group D than in group C at 6 months after operation ( P<0.05). HE staining showed that there were significantly more new tissues in the middle of the nerve conduit in group D after operation than in other groups. Immunohistochemical staining showed that CD86 and CD206 stainings were positive in each group at 1 month after operation, among which group D had the lowest positive rate of CD86 and the highest positive rate of CD206, and there were significant differences in the positive rate of CD206 between group D and groups A, B, and C ( P<0.05); the MBP and P0 stainings were positive in groups C and D at 6 months, and the positive rate in group D was significantly higher than that in group C ( P<0.05). Immunofluorescence staining showed that the NF-200 and S-100β expressions in group D were significantly higher than those in other groups. CONCLUSION Telmisartan/collagen/PLC nerve conduit can promote the sciatic nerve defect repair in rats through promoting the polarization of M1 macrophages to M2 macrophages, and the nerve conduit loaded with20 mg telmisartan has the most significant effect.
Collapse
|
11
|
Hu W, Wang Y, Chen J, Yu P, Tang F, Hu Z, Zhou J, Liu L, Qiu W, Ye Y, Jia Y, Zhou S, Long J, Zeng Z. Regulation of biomaterial implantation-induced fibrin deposition to immunological functions of dendritic cells. Mater Today Bio 2022; 14:100224. [PMID: 35252832 PMCID: PMC8894278 DOI: 10.1016/j.mtbio.2022.100224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 11/04/2022] Open
Abstract
The performance of implanted biomaterials is largely determined by their interaction with the host immune system. As a fibrous-like 3D network, fibrin matrix formed at the interfaces of tissue and material, whose effects on dendritic cells (DCs) remain unknown. Here, a bone plates implantation model was developed to evaluate the fibrin matrix deposition and DCs recruitment in vivo. The DCs responses to fibrin matrix were further analyzed by a 2D and 3D fibrin matrix model in vitro. In vivo results indicated that large amount of fibrin matrix deposited on the interface between the tissue and bone plates, where DCs were recruited. Subsequent in vitro testing denoted that DCs underwent significant shape deformation and cytoskeleton reorganization, as well as mechanical property alteration. Furthermore, the immune function of imDCs and mDCs were negatively and positively regulated, respectively. The underlying mechano-immunology coupling mechanisms involved RhoA and CDC42 signaling pathways. These results suggested that fibrin plays a key role in regulating DCs immunological behaviors, providing a valuable immunomodulatory strategy for tissue healing, regeneration and implantation.
Collapse
|
12
|
Russo V, El Khatib M, Prencipe G, Cerveró-Varona A, Citeroni MR, Mauro A, Berardinelli P, Faydaver M, Haidar-Montes AA, Turriani M, Di Giacinto O, Raspa M, Scavizzi F, Bonaventura F, Liverani L, Boccaccini AR, Barboni B. Scaffold-Mediated Immunoengineering as Innovative Strategy for Tendon Regeneration. Cells 2022; 11:cells11020266. [PMID: 35053383 PMCID: PMC8773518 DOI: 10.3390/cells11020266] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Tendon injuries are at the frontier of innovative approaches to public health concerns and sectoral policy objectives. Indeed, these injuries remain difficult to manage due to tendon’s poor healing ability ascribable to a hypo-cellularity and low vascularity, leading to the formation of a fibrotic tissue affecting its functionality. Tissue engineering represents a promising solution for the regeneration of damaged tendons with the aim to stimulate tissue regeneration or to produce functional implantable biomaterials. However, any technological advancement must take into consideration the role of the immune system in tissue regeneration and the potential of biomaterial scaffolds to control the immune signaling, creating a pro-regenerative environment. In this context, immunoengineering has emerged as a new discipline, developing innovative strategies for tendon injuries. It aims at designing scaffolds, in combination with engineered bioactive molecules and/or stem cells, able to modulate the interaction between the transplanted biomaterial-scaffold and the host tissue allowing a pro-regenerative immune response, therefore hindering fibrosis occurrence at the injury site and guiding tendon regeneration. Thus, this review is aimed at giving an overview on the role exerted from different tissue engineering actors in leading immunoregeneration by crosstalking with stem and immune cells to generate new paradigms in designing regenerative medicine approaches for tendon injuries.
Collapse
Affiliation(s)
- Valentina Russo
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Mohammad El Khatib
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
- Correspondence:
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maria Rita Citeroni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Paolo Berardinelli
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Melisa Faydaver
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Arlette A. Haidar-Montes
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maura Turriani
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Oriana Di Giacinto
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Marcello Raspa
- Institute of Biochemistry and Cellular Biology (IBBC), Council of National Research (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Ferdinando Scavizzi
- Institute of Biochemistry and Cellular Biology (IBBC), Council of National Research (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Fabrizio Bonaventura
- Institute of Biochemistry and Cellular Biology (IBBC), Council of National Research (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Liliana Liverani
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (L.L.); (A.R.B.)
| | - Aldo R. Boccaccini
- Department of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (L.L.); (A.R.B.)
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (A.C.-V.); (M.R.C.); (A.M.); (P.B.); (M.F.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| |
Collapse
|
13
|
Cao L, Tong Y, Wang X, Zhang Q, Qi Y, Zhou C, Yu X, Wu Y, Miao X. Effect of Amniotic Membrane/Collagen-Based Scaffolds on the Chondrogenic Differentiation of Adipose-Derived Stem Cells and Cartilage Repair. Front Cell Dev Biol 2021; 9:647166. [PMID: 34900977 PMCID: PMC8657407 DOI: 10.3389/fcell.2021.647166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 10/20/2021] [Indexed: 11/24/2022] Open
Abstract
Objectives: Repairing articular cartilage damage is challenging. Clinically, tissue engineering technology is used to induce stem cell differentiation and proliferation on biological scaffolds to repair defective joints. However, no ideal biological scaffolds have been identified. This study investigated the effects of amniotic membrane/collagen scaffolds on the differentiation of adipose-derived stem cells (ADSCs) and articular cartilage repair. Methods: Adipose tissue of New Zealand rabbits was excised, and ADSCs were isolated and induced for differentiation. An articular cartilage defect model was constructed to identify the effect of amniotic membrane/collagen scaffolds on cartilage repair. Cartilage formation was analyzed by imaging and toluene blue staining. Knee joint recovery in rabbits was examined using hematoxylin and eosin, toluidine, safranine, and immunohistochemistry at 12 weeks post-operation. Gene expression was examined using ELISA, RT-PCR, Western blotting, and immunofluorescence. Results: The adipose tissue was effectively differentiated into ADSCs, which further differentiated into chondrogenic, osteogenic, and lipogenic lineages after 3 weeks’ culture in vitro. Compared with platelet-rich plasmon (PRP) scaffolds, the amniotic membrane scaffolds better promoted the growth and differentiation of ADSCs. Additionally, scaffolds containing the PRP and amniotic membrane efficiently enhanced the osteogenic differentiation of ADSCs. The levels of COL1A1, COL2A1, COL10A1, SOX9, and ACAN in ADSCs + amniotic membrane + PRP group were significantly higher than the other groups both in vitro and in vivo. The Wakitani scores of the ADSC + amniotic membrane + PRP group were lower than that in ADSC + PRP (4.4 ± 0.44**), ADSC + amniotic membrane (2.63 ± 0.38**), and control groups (6.733 ± 0.21) at week 12 post-operation. Osteogenesis in rabbits of the ADSC + amniotic membrane + PRP group was significantly upregulated when compared with other groups. Amniotic membranes significantly promoted the expression of cartilage regeneration-related factors (SOX6, SOX9, RUNX2, NKX3-2, MEF2C, and GATA4). The ADSC + PRP + amniotic membrane group exhibited the highest levels of TGF-β, PDGF, and FGF while exhibiting the lowest level of IL-1β, IL6, and TNF-α in articular cavity. Conclusion: Amniotic membrane/collagen combination-based scaffolds promoted the proliferation and cartilage differentiation of ADSCs, and may provide a new treatment paradigm for patients with cartilage injury.
Collapse
Affiliation(s)
- Le Cao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Yuling Tong
- Department of General Practice, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Wang
- Shaoxing Shangyu Hospital of Traditional Chinese medicine, Shaoxing, China
| | - Qiang Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Yiying Qi
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Chenhe Zhou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Xinning Yu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Yongping Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Xudong Miao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| |
Collapse
|
14
|
Zhu G, Zhang T, Chen M, Yao K, Huang X, Zhang B, Li Y, Liu J, Wang Y, Zhao Z. Bone physiological microenvironment and healing mechanism: Basis for future bone-tissue engineering scaffolds. Bioact Mater 2021; 6:4110-4140. [PMID: 33997497 PMCID: PMC8091181 DOI: 10.1016/j.bioactmat.2021.03.043] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/19/2021] [Accepted: 03/28/2021] [Indexed: 02/06/2023] Open
Abstract
Bone-tissue defects affect millions of people worldwide. Despite being common treatment approaches, autologous and allogeneic bone grafting have not achieved the ideal therapeutic effect. This has prompted researchers to explore novel bone-regeneration methods. In recent decades, the development of bone tissue engineering (BTE) scaffolds has been leading the forefront of this field. As researchers have provided deep insights into bone physiology and the bone-healing mechanism, various biomimicking and bioinspired BTE scaffolds have been reported. Now it is necessary to review the progress of natural bone physiology and bone healing mechanism, which will provide more valuable enlightenments for researchers in this field. This work details the physiological microenvironment of the natural bone tissue, bone-healing process, and various biomolecules involved therein. Next, according to the bone physiological microenvironment and the delivery of bioactive factors based on the bone-healing mechanism, it elaborates the biomimetic design of a scaffold, highlighting the designing of BTE scaffolds according to bone biology and providing the rationale for designing next-generation BTE scaffolds that conform to natural bone healing and regeneration.
Collapse
Affiliation(s)
- Guanyin Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Tianxu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Miao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Ke Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Xinqi Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Bo Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Yazhen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Jun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| |
Collapse
|
15
|
Mei X, Ye D, Zhang F, Di C. Implantable application of polymer‐based biosensors. JOURNAL OF POLYMER SCIENCE 2021. [DOI: 10.1002/pol.20210543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Xiangyuan Mei
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids Institute of Chemistry, Chinese Academy of Sciences Beijing China
- School of Chemical Sciences University of Chinese Academy of Sciences Beijing China
| | - Dekai Ye
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids Institute of Chemistry, Chinese Academy of Sciences Beijing China
| | - Fengjiao Zhang
- School of Chemical Sciences University of Chinese Academy of Sciences Beijing China
| | - Chong‐an Di
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids Institute of Chemistry, Chinese Academy of Sciences Beijing China
| |
Collapse
|
16
|
Chen L, Zhang L, Zhang H, Sun X, Liu D, Zhang J, Zhang Y, Cheng L, Santos HA, Cui W. Programmable immune activating electrospun fibers for skin regeneration. Bioact Mater 2021; 6:3218-3230. [PMID: 33778200 PMCID: PMC7966852 DOI: 10.1016/j.bioactmat.2021.02.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/02/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
Immune cells play a crucial regulatory role in inflammatory phase and proliferative phase during skin healing. How to programmatically activate sequential immune responses is the key for scarless skin regeneration. In this study, an "Inner-Outer" IL-10-loaded electrospun fiber with cascade release behavior was constructed. During the inflammatory phase, the electrospun fiber released a lower concentration of IL-10 within the wound, inhibiting excessive recruitment of inflammatory cells and polarizing macrophages into anti-inflammatory phenotype "M2c" to suppress excessive inflammation response. During the proliferative phase, a higher concentration of IL-10 released by the fiber and the anti-fibrotic cytokines secreted by polarized "M2c" directly acted on dermal fibroblasts to simultaneously inhibit extracellular matrix overdeposition and promote fibroblast migration. The "Inner-Outer" IL-10-loaded electrospun fiber programmatically activated the sequential immune responses during wound healing and led to scarless skin regeneration, which is a promising immunomodulatory biomaterial with great potential for promoting complete tissue regeneration.
Collapse
Affiliation(s)
- Lu Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai 200011, PR China
| | - Liucheng Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai 200011, PR China
| | - Hongbo Zhang
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China.,Department of Pharmaceutical Sciences Laboratory and Turku Center for Biotechnology, Åbo Akademi University, Turku FI-20520, Finland
| | - Xiaoming Sun
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai 200011, PR China
| | - Dan Liu
- National Research Center for Translational Medicine, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Jianming Zhang
- National Research Center for Translational Medicine, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Yuguang Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai 200011, PR China
| | - Liying Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai 200011, PR China
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland.,Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki FI-00014, Finland
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| |
Collapse
|
17
|
Zhang J, Shi H, Zhang N, Hu L, Jing W, Pan J. Interleukin-4-loaded hydrogel scaffold regulates macrophages polarization to promote bone mesenchymal stem cells osteogenic differentiation via TGF-β1/Smad pathway for repair of bone defect. Cell Prolif 2020; 53:e12907. [PMID: 32951298 PMCID: PMC7574882 DOI: 10.1111/cpr.12907] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/18/2020] [Accepted: 08/31/2020] [Indexed: 02/05/2023] Open
Abstract
Objective Tissue engineering is a promising strategy for repair of large bone defect. However, the immune system reactions to biological scaffold are increasingly being recognized as a crucial factor influencing regeneration efficacy. In this study, a bone‐bioactive hydrogel bead loaded with interleukin‐4 (IL‐4) was used to regulate macrophages polarization and accelerate bone regeneration. Methods IL‐4‐loaded calcium‐enriched gellan gum (Ca‐GG + IL‐4) hydrogel beads were synthesised. And the effect on cell behaviour was detected. Furthermore, the effect of the Ca‐GG + IL‐4 hydrogel bead on macrophage polarization and the effect of macrophage polarization on bone mesenchymal stem cells (BMSCs) apoptosis and osteogenic differentiation were evaluated in vitro and in vivo. Results BMSCs were able to survive in the hydrogel regardless of whether IL‐4 was incorporated. Immunofluorescence staining and qPCR results revealed that Ca‐GG + IL‐4 hydrogel bead could promote M2 macrophage polarization and increase transforming growth factor (TGF)‐β1 expression level, which activates the TGF‐β1/Smad signalling pathway in BMSCs and promotes osteogenic differentiation. Moreover, immunohistochemical analysis demonstrated Ca‐GG + IL‐4 hydrogel bead could promote M2 macrophage polarization and reduce cell apoptosis in vivo. In addition, micro‐CT and immunohistochemical analysis at 12 weeks post‐surgery showed that Ca‐GG + IL‐4 hydrogel bead could achieve superior bone defect repair efficacy in vivo. Conclusions The Ca‐GG + IL‐4 hydrogel bead effectively promoted bone defect regeneration via regulating macrophage polarization, reducing cell apoptosis and promoting BMSCs osteogenesis through TGF‐β1/Smad pathway. Therefore, it is a promising strategy for repair of bone defect.
Collapse
Affiliation(s)
- Jiankang Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haitao Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Nian Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liru Hu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Higuchi J, Fortunato G, Woźniak B, Chodara A, Domaschke S, Męczyńska-Wielgosz S, Kruszewski M, Dommann A, Łojkowski W. Polymer Membranes Sonocoated and Electrosprayed with Nano-Hydroxyapatite for Periodontal Tissues Regeneration. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1625. [PMID: 31731775 PMCID: PMC6915502 DOI: 10.3390/nano9111625] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 01/21/2023]
Abstract
Diseases of periodontal tissues are a considerable clinical problem, connected with inflammatory processes and bone loss. The healing process often requires reconstruction of lost bone in the periodontal area. For that purpose, various membranes are used to prevent ingrowth of epithelium in the tissue defect and enhance bone regeneration. Currently-used membranes are mainly non-resorbable or are derived from animal tissues. Thus, there is an urgent need for non-animal-derived bioresorbable membranes with tuned resorption rates and porosity optimized for the circulation of body nutrients. We demonstrate membranes produced by the electrospinning of biodegradable polymers (PDLLA/PLGA) coated with nanohydroxyapatite (nHA). The nHA coating was made using two methods: sonocoating and electrospraying of nHA suspensions. In a simulated degradation study, for electrosprayed membranes, short-term calcium release was observed, followed by hydrolytic degradation. Sonocoating produced a well-adhering nHA layer with full coverage of the fibers. The layer slowed the polymer degradation and increased the membrane wettability. Due to gradual release of calcium ions the degradation-associated acidity of the polymer was neutralized. The sonocoated membranes exhibited good cellular metabolic activity responses against MG-63 and BJ cells. The collected results suggest their potential use in Guided Tissue Regeneration (GTR) and Guided Bone Regeneration (GBR) periodontal procedures.
Collapse
Affiliation(s)
- Julia Higuchi
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, 01142 Warsaw, Poland; (B.W.); (A.C.); (W.Ł.)
- Faculty of Materials Science and Engineering, Warsaw University of Technology, 02507 Warsaw, Poland
- Laboratory for Biomimetic Membranes and Textiles, Empa Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland
| | - Giuseppino Fortunato
- Laboratory for Biomimetic Membranes and Textiles, Empa Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland
| | - Bartosz Woźniak
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, 01142 Warsaw, Poland; (B.W.); (A.C.); (W.Ł.)
| | - Agnieszka Chodara
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, 01142 Warsaw, Poland; (B.W.); (A.C.); (W.Ł.)
- Faculty of Materials Science and Engineering, Warsaw University of Technology, 02507 Warsaw, Poland
| | - Sebastian Domaschke
- Experimental Continuum Mechanics, Empa Swiss Federal Laboratories for Materials Science and Technology, 8600 Dübendorf, Switzerland;
- Department of Mechanical and Process Engineering, Institute for Mechanical Systems, ETH Zürich, 8092 Zürich, Switzerland
| | - Sylwia Męczyńska-Wielgosz
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, 03195 Warsaw, Poland;
| | - Marcin Kruszewski
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 20090 Lublin, Poland;
| | - Alex Dommann
- Department Materials meet Life, Empa Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland;
| | - Witold Łojkowski
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, 01142 Warsaw, Poland; (B.W.); (A.C.); (W.Ł.)
| |
Collapse
|
19
|
Yang D, Xiao J, Wang B, Li L, Kong X, Liao J. The immune reaction and degradation fate of scaffold in cartilage/bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109927. [DOI: 10.1016/j.msec.2019.109927] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/17/2019] [Accepted: 06/26/2019] [Indexed: 01/05/2023]
|
20
|
Sutter D, Dzhonova DV, Prost JC, Bovet C, Banz Y, Rahnfeld L, Leroux JC, Rieben R, Vögelin E, Plock JA, Luciani P, Taddeo A, Schnider JT. Delivery of Rapamycin Using In Situ Forming Implants Promotes Immunoregulation and Vascularized Composite Allograft Survival. Sci Rep 2019; 9:9269. [PMID: 31239498 PMCID: PMC6592945 DOI: 10.1038/s41598-019-45759-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 06/13/2019] [Indexed: 12/11/2022] Open
Abstract
Vascularized composite allotransplantation (VCA), such as hand and face transplantation, is emerging as a potential solution in patients that suffered severe injuries. However, adverse effects of chronic high-dose immunosuppression regimens strongly limit the access to these procedures. In this study, we developed an in situ forming implant (ISFI) loaded with rapamycin to promote VCA acceptance. We hypothesized that the sustained delivery of low-dose rapamycin in proximity to the graft may promote graft survival and induce an immunoregulatory microenvironment, boosting the expansion of T regulatory cells (Treg). In vitro and in vivo analysis of rapamycin-loaded ISFI (Rapa-ISFI) showed sustained drug release with subtherapeutic systemic levels and persistent tissue levels. A single injection of Rapa-ISFI in the groin on the same side as a transplanted limb significantly prolonged VCA survival. Moreover, treatment with Rapa-ISFI increased the levels of multilineage mixed chimerism and the frequency of Treg both in the circulation and VCA-skin. Our study shows that Rapa-ISFI therapy represents a promising approach for minimizing immunosuppression, decreasing toxicity and increasing patient compliance. Importantly, the use of such a delivery system may favor the reprogramming of allogeneic responses towards a regulatory function in VCA and, potentially, in other transplants and inflammatory conditions.
Collapse
Affiliation(s)
- Damian Sutter
- Department of Plastic and Hand Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Jean-Christophe Prost
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Cedric Bovet
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yara Banz
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Lisa Rahnfeld
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Jena, Jena, Germany.,Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Robert Rieben
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Esther Vögelin
- Department of Plastic and Hand Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Jan A Plock
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, University of Zurich, Zürich, Switzerland.
| | - Paola Luciani
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Jena, Jena, Germany. .,Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland. .,Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland.
| | - Adriano Taddeo
- Department of Plastic and Hand Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland. .,Department for BioMedical Research, University of Bern, Bern, Switzerland.
| | - Jonas T Schnider
- Department of Plastic and Hand Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
21
|
Wang C, Chen B, Wang W, Zhang X, Hu T, He Y, Lin K, Liu X. Strontium released bi-lineage scaffolds with immunomodulatory properties induce a pro-regenerative environment for osteochondral regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109833. [PMID: 31349499 DOI: 10.1016/j.msec.2019.109833] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/13/2019] [Accepted: 05/29/2019] [Indexed: 02/07/2023]
Abstract
The different lineage-specific biological properties of articular cartilage and subchondral bone present a great challenge in the construction of bi-lineage scaffolds for simultaneous osteochondral regeneration. To overcome this challenge, strontium incorporated calcium silicate (Sr-CS) ceramic was prepared for bi-lineage formation of scaffolds in this study. The positive result of Sr-CS in the regeneration of osteochondral defects was first proven by its improved effect on the osteogenesis and chondrogenesis induction of mesenchymal stem cells (MSCs). After that, scaffold-mediated macrophage polarization between classically activated inflammatory macrophages (termed M1Ф) and alternatively activated inflammatory macrophages (termed M2Ф) was assayed to investigate whether the incorporation of Sr into calcium silicate could alter host-to-scaffold immune response. Furthermore, the interactions between Sr-CS pretreated macrophages and MSCs differentiation were performed to prove the enhancement effect of suppressed inflammatory response on osteogenesis and chondrogenesis. In vivo transplantation showed that the Sr-CS scaffolds distinctly improved the regeneration of cartilage and subchondral bone, as compared to the calcium silicate scaffolds. On the one hand, the mechanism attributes to enhancement of strontium on the osteogenic and chondrogenic differentiation of MSCs. On the other hand, the reason can partially be attributed to suppressed synovial inflammatory response, which has improved effects on enhancement of osteogenesis and chondrogenesis. These findings suggest that monophasic Sr-CS scaffolds with a bi-lineage conducive property and an inflammatory response regulatory property represents a viable strategy for simultaneous regeneration of osteochondral defects.
Collapse
Affiliation(s)
- Chongyang Wang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Bi Chen
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Wei Wang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital East Campus, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China; Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Xuancheng Zhang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Tu Hu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Yaohua He
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Kaili Lin
- Department of Oral and Cranio-maxillofacial Science, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China.
| | - Xudong Liu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.
| |
Collapse
|
22
|
Biomaterials: Foreign Bodies or Tuners for the Immune Response? Int J Mol Sci 2019; 20:ijms20030636. [PMID: 30717232 PMCID: PMC6386828 DOI: 10.3390/ijms20030636] [Citation(s) in RCA: 363] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 12/11/2022] Open
Abstract
The perspectives of regenerative medicine are still severely hampered by the host response to biomaterial implantation, despite the robustness of technologies that hold the promise to recover the functionality of damaged organs and tissues. In this scenario, the cellular and molecular events that decide on implant success and tissue regeneration are played at the interface between the foreign body and the host inflammation, determined by innate and adaptive immune responses. To avoid adverse events, rather than the use of inert scaffolds, current state of the art points to the use of immunomodulatory biomaterials and their knowledge-based use to reduce neutrophil activation, and optimize M1 to M2 macrophage polarization, Th1 to Th2 lymphocyte switch, and Treg induction. Despite the fact that the field is still evolving and much remains to be accomplished, recent research breakthroughs have provided a broader insight on the correct choice of biomaterial physicochemical modifications to tune the reaction of the host immune system to implanted biomaterial and to favor integration and healing.
Collapse
|
23
|
Li YM, Wu JY, Jiang J, Dong SK, Chen YS, He HY, Liu CS, Zhao JZ. Chondroitin sulfate-polydopamine modified polyethylene terephthalate with extracellular matrix-mimetic immunoregulatory functions for osseointegration. J Mater Chem B 2019; 7:7756-7770. [DOI: 10.1039/c9tb01984g] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Optimal integration between the polyethylene terephthalate (PET) graft and host bone is a prerequisite to obtain a satisfactory outcome after graft implantation for ligament reconstruction.
Collapse
Affiliation(s)
- Ya-Min Li
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Jing-Yao Wu
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai
- China
| | - Jia Jiang
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Shi-Kui Dong
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Yun-Su Chen
- Department of Joint Surgery
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Hong-Yan He
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai
- China
| | - Chang-Sheng Liu
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai
- China
| | - Jin-Zhong Zhao
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| |
Collapse
|
24
|
Carpenter RA, Kwak JG, Peyton SR, Lee J. Implantable pre-metastatic niches for the study of the microenvironmental regulation of disseminated human tumour cells. Nat Biomed Eng 2018; 2:915-929. [PMID: 30906645 PMCID: PMC6424369 DOI: 10.1038/s41551-018-0307-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 09/06/2018] [Indexed: 02/07/2023]
Abstract
Cancer survivors often carry disseminated tumour cells (DTCs), yet owing to DTC dormancy they do not relapse from treatment. Understanding how the local microenvironment regulates the transition of DTCs from a quiescent state to active proliferation could suggest new therapeutic strategies to prevent or delay the formation of metastases. Here, we show that implantable biomaterial microenvironments incorporating human stromal cells, immune cells and cancer cells can be used to examine the post-dissemination phase of the evolution of the tumour microenvironment. After subdermal implantation in mice, porous hydrogel scaffolds seeded with human bone marrow stromal cells form a vascularized niche and recruit human circulating tumour cells released from an orthotopic prostate tumour xenograft. Systemic injection of human peripheral blood mononuclear cells slowed the evolution of the active metastatic niches but did not change the rate of overt metastases, as the ensuing inflammation promoted the formation of DTC colonies. Implantable pre-metastatic niches might enable the study of DTC colonization and proliferation, and facilitate the development of effective anti-metastatic therapies.
Collapse
Affiliation(s)
- Ryan A Carpenter
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| | - Jun-Goo Kwak
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| | - Shelly R Peyton
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA
| | - Jungwoo Lee
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA.
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
25
|
Zhu M, Wu Y, Li W, Dong X, Chang H, Wang K, Wu P, Zhang J, Fan G, Wang L, Liu J, Wang H, Kong D. Biodegradable and elastomeric vascular grafts enable vascular remodeling. Biomaterials 2018; 183:306-318. [DOI: 10.1016/j.biomaterials.2018.08.063] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 02/07/2023]
|
26
|
Menale C, Campodoni E, Palagano E, Mantero S, Erreni M, Inforzato A, Fontana E, Schena F, Van't Hof R, Sandri M, Tampieri A, Villa A, Sobacchi C. Mesenchymal Stromal Cell-Seeded Biomimetic Scaffolds as a Factory of Soluble RANKL in Rankl-Deficient Osteopetrosis. Stem Cells Transl Med 2018; 8:22-34. [PMID: 30184340 PMCID: PMC6312453 DOI: 10.1002/sctm.18-0085] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/11/2018] [Indexed: 12/27/2022] Open
Abstract
Biomimetic scaffolds are extremely versatile in terms of chemical composition and physical properties, which can be defined to accomplish specific applications. One property that can be added is the production/release of bioactive soluble factors, either directly from the biomaterial, or from cells embedded within the biomaterial. We reasoned that pursuing this strategy would be appropriate to setup a cell‐based therapy for RANKL‐deficient autosomal recessive osteopetrosis, a very rare skeletal genetic disease in which lack of the essential osteoclastogenic factor RANKL impedes osteoclast formation. The exogenously administered RANKL cytokine is effective in achieving osteoclast formation and function in vitro and in vivo, thus, we produced murine Rankl−/− mesenchymal stromal cells (MSCs) overexpressing human soluble RANKL (hsRL) following lentiviral transduction (LVhsRL). Here, we described a three‐dimensional (3D) culture system based on a magnesium‐doped hydroxyapatite/collagen I (MgHA/Col) biocompatible scaffold closely reproducing bone physicochemical properties. MgHA/Col‐seeded murine MSCs showed improved properties, as compared to two‐dimensional (2D) culture, in terms of proliferation and hsRL production, with respect to LVhsRL‐transduced cells. When implanted subcutaneously in Rankl−/− mice, these cell constructs were well tolerated, colonized by host cells, and intensely vascularized. Of note, in the bone of Rankl−/− mice that carried scaffolds with either WT or LVhsRL‐transduced Rankl−/− MSCs, we specifically observed formation of TRAP+ cells, likely due to sRL released from the scaffolds into circulation. Thus, our strategy proved to have the potential to elicit an effect on the bone; further work is required to maximize these benefits and achieve improvements of the skeletal pathology in the treated Rankl−/− mice. Stem Cells Translational Medicine2019;8:22–34
Collapse
Affiliation(s)
- Ciro Menale
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | | | - Eleonora Palagano
- Humanitas Clinical and Research Institute, Rozzano, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Stefano Mantero
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Marco Erreni
- Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Antonio Inforzato
- Humanitas Clinical and Research Institute, Rozzano, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Elena Fontana
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Francesca Schena
- Clinica Pediatrica e Reumatologia, UOSD Centro Malattie Autoinfiammatorie e Immunodeficienze, Genoa, Italy
| | - Rob Van't Hof
- Bone Research Group, Institute of Ageing & Chronic Disease, University of Liverpool, Liverpool, UK
| | | | | | - Anna Villa
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| | - Cristina Sobacchi
- CNR-IRGB, Milan Unit, Milan, Italy.,Humanitas Clinical and Research Institute, Rozzano, Italy
| |
Collapse
|
27
|
Abstract
Purpose The focus of this review is to provide an overview of the recent findings on the role of epigenetic mechanisms in periodontal disease, including disease susceptibility, progression, and as potential treatment options. Recent Findings The findings on the influence of oral pathogens on epigenetic regulation of pathogen recognition receptors, such as Toll-like receptors, as well as pro-inflammatory cytokines suggest an important role for epigenetics in the regulation of the host immune response. Recent studies also show that the epigenetic pattern in periodontitis lesions differ from that of healthy and gingivitis tissue. In addition, these patterns differ between tissues in the same individual. Research is also indicating a role for both DNA methylation and histone acetylation on cells osteogenic differentiation and bone regeneration. Summary Knowledge of epigenetic pattern in periodontal diseases may add not only to the knowledge of susceptibility of the disease but may also be a diagnostic tool to identify patients at risk to develop the severe form of periodontitis. In addition, recent research within gene therapy and tissue engineering indicate a role for epigenetics also to improve regeneration of periodontal tissues.
Collapse
Affiliation(s)
- Lena Larsson
- Department of Periodontology, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Box 450, SE-405 30 Gothenburg, Sweden
| |
Collapse
|