1
|
Feng J, Wang F, Shao Y, Jin A, Lei L. Engineered protein-based materials for tissue repair: A review. Int J Biol Macromol 2025; 303:140674. [PMID: 39909268 DOI: 10.1016/j.ijbiomac.2025.140674] [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: 12/09/2024] [Revised: 01/19/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
The human body may suffer multiple injuries and losses due to various external factors, such as tumors, diseases, traffic accidents, and war conflicts. Under such circumstances, engineered protein-based materials, as an innovative adjunctive material, can not only effectively promote the natural repair process of tissues, but also greatly circumvent the negative effects and complications that may be associated with conventional surgery. In this review, we first trace the definition and development of engineered protein-based materials and explain in detail their mechanism of action in promoting tissue repair. Subsequently, the advantages and disadvantages of various engineered protein-based materials in tissue repair are analyzed by comparison. In addition, the present review reveals in depth how material properties can be optimized by scientific means to meet different tissue repair needs. In addition, we present in detail specific application cases of engineered protein-based materials in the field of tissue repair. Finally, we summarize current challenges in engineered protein-based materials and provide an outlook for the future. This review not only provides theoretical support for the further exploration and development of engineered protein-based materials in the field of tissue repair, but also provides valuable references and inspiration for research in related fields.
Collapse
Affiliation(s)
- Jiayin Feng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Fangyan Wang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Yunyuan Shao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Anqi Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China.
| |
Collapse
|
2
|
Pulat G, Bilgiç E, Ercan UK, Karaman O. Enhanced Osteogenic Differentiation via Collagen and BMP-2 Mimetic Peptide Conjugation to β-TCP Scaffolds Using a Cold Atmospheric Plasma-Assisted Strategy. ACS APPLIED BIO MATERIALS 2025; 8:2569-2579. [PMID: 40032828 DOI: 10.1021/acsabm.5c00029] [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: 03/05/2025]
Abstract
Bone defects arising from trauma, disease, or surgical intervention represent significant challenges. Developing effective bone tissue engineering strategies to address these issues and promote repair is crucial. β-Tricalcium phosphate (β-TCP) has emerged as a promising synthetic graft due to its porous, degradable structure and excellent biocompatibility. However, the lack of biological cues in β-TCP limits its functionality, requiring different surface modification strategies. Bone morphogenetic protein-2 mimetic peptide (BMP; NSVNSKIPKACCVPTELSAI) and collagen mimetic peptide (CMP; GTPGPQGIAGQRGVV) have a known significant therapeutic potential due to their ability to enhance cell attachment and osteogenic differentiation. Herein, a peptide functionalization strategy for β-TCP scaffolds was introduced. Briefly, β-TCP was treated with cold atmospheric plasma (CAP) to create functional hydroxyl groups on the surface of the β-TCP. Subsequently, peptides were conjugated by using a three-step method: (1) silanization with APTES, (2) EDC activation, and (3) peptide conjugation. The successful surface modification with CAP and peptide conjugation was confirmed via XRD, FTIR, and Raman analysis. Furthermore, the effects of BMP and CMP peptides on osteogenic differentiation after CAP treatment were investigated in human mesenchymal stem cells (hMSCs). Both β-TCP/BMP and β-TCP/CMP scaffolds demonstrated excellent biocompatibility with hMSCs, enhancing cell proliferation and promoting osteogenic differentiation. Remarkably, β-TCP/CMP showed better results in terms of proliferation and differentiation compared with β-TCP/BMP. These findings highlight the clinical potential of peptide-functionalized β-TCP scaffolds for bone tissue engineering while also providing a promising methodology for β-TCP functionalization.
Collapse
Affiliation(s)
- Günnur Pulat
- Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir 35620, Turkey
| | - Eda Bilgiç
- Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir 35620, Turkey
| | - Utku Kürşat Ercan
- Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir 35620, Turkey
| | - Ozan Karaman
- Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir 35620, Turkey
| |
Collapse
|
3
|
Artamonov MY, Pyatakovich FA, Minenko IA. Synergistic Antioxidant Effects of Molecular Hydrogen and Cold Atmospheric Plasma in Enhancing Mesenchymal Stem Cell Therapy. Antioxidants (Basel) 2024; 13:1584. [PMID: 39765910 PMCID: PMC11673711 DOI: 10.3390/antiox13121584] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
In regenerative medicine, mesenchymal stem cells (MSCs) have shown their importance and potential in tissue reconstruction and immune system modification. However, such cells' potential is often diminished by factors such as oxidative stress, immune rejection, and inadequate engraftment. This review highlights the role of molecular hydrogen (H2) and cold atmospheric plasma (CAP) as adjunct therapies to improve the effectiveness of MSC therapy. H2 has strong antioxidative and anti-inflammatory actions as it quenches reactive oxygen species and positively stimulates the Nrf2 pathway that promotes MSC survival and life. CAP, being a modulated source of ROS and RNS, also assists MSCs by altering the cellular redox balance, thus facilitating cellular adaptation, migration, and differentiation. H2 and CAP in conjunction with each other assist in establishing an ambience favorable for promoting MSCs' survival and growth abilities, and reduce the healing time in various pathways such as wound, neuroprotection, and ischemia. Besides these concerns, this review also covers the best administration routes and doses of H2 and CAP together with MSCs in therapy. This study informs on a novel dual method aimed at improving the outcome of MSC therapy while adding several molecular targets and relevant clinical uses concerning these therapies. Research of the future has to deal with bettering these protocols so that the therapeutic benefits can be maximized without long-term implications for clinical applications.
Collapse
Affiliation(s)
- Mikhail Yu. Artamonov
- Department of Physical Medicine and Rehabilitation, Penn Medicine Princeton Health, Plainsboro, NJ 08536, USA
| | - Felix A. Pyatakovich
- Department of Internal Medicine, Belgorod State University, 308015 Belgorod, Russia;
| | - Inessa A. Minenko
- Department of Rehabilitation, Sechenov Medical University, 119991 Moscow, Russia;
| |
Collapse
|
4
|
Ortea I, Rodríguez-Martínez L, Carrera M, Fafián-Labora JA, Arufe MC, González-Barcia M, Fernández-Ferreiro A, Mateos J. ZenoSWATH DIA proteomics and clustering analysis of the effect of cysteamine at the cellular level in cystinotic fibroblasts. Biomed Pharmacother 2024; 181:117650. [PMID: 39504626 DOI: 10.1016/j.biopha.2024.117650] [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: 08/05/2024] [Revised: 10/14/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Cysteamine, an aminothiol, is the only available treatment for cystinosis, an incurable metabolic recessive disease characterized by detrimental symptoms at the renal, ocular, and muscular levels. Cystinosis is due to mutations in the CTNS gene encoding for the lysosomal symporter cystinosine. Cysteamine treatment only delays the symptoms, presents undesirable side effects and the patients depend on it for life. Thus, it is of paramount importance to find new complementary therapeutic targets for the disease, as well as to understand, at the molecular level, both the beneficial and detrimental effects of cysteamine. Here, we have used ZenoSWATH DIA proteomics and clustering analysis to unravel the differences between cystinotic and non-cystinotic skin fibroblasts, and to study the effect of increasing concentrations of cysteamine. Cystinotic cells present significant differences in proteins related to extracellular matrix structure and detoxification. Only a subset of those proteins is reversed by cysteamine in a dose-dependent manner, partially providing an explanation for its therapeutic benefits. Finally, cysteamine per se alters the levels of a group of lysosomal proteins that are not modulated in basal conditions. Our results will be helpful to understand the benefits, deficiencies, and detrimental effects of the cysteamine treatment.
Collapse
Affiliation(s)
- Ignacio Ortea
- Proteomics Unit, Centro de Investigación en Nanomateriales y Nanotecnología (CINN-CSIC), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| | - Lorena Rodríguez-Martínez
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Mónica Carrera
- Institute of Marine Research (IIM), Spanish National Research Council (CSIC), Vigo 36208, Spain
| | - Juan A Fafián-Labora
- Grupo de Terapia Celular y Medicina Regenerativa, Dpto. de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de La Salud, Universidade da Coruña, INIBIC, CHUAC, CICA, A Coruña 15006, Spain
| | - Maria C Arufe
- Grupo de Terapia Celular y Medicina Regenerativa, Dpto. de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de La Salud, Universidade da Coruña, INIBIC, CHUAC, CICA, A Coruña 15006, Spain
| | - Miguel González-Barcia
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Anxo Fernández-Ferreiro
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
| | - Jesús Mateos
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
| |
Collapse
|
5
|
Ogiwara N, Nakano T, Baba K, Noguchi H, Masuda T, Takai M. High-Quality Three-Dimensionally Cultured Cells Using Interfaces of Diblock Copolymers Containing Different Ratios of Zwitterionic N-Oxides. ACS APPLIED MATERIALS & INTERFACES 2024; 16:44575-44589. [PMID: 39160767 PMCID: PMC11368093 DOI: 10.1021/acsami.4c10118] [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: 06/18/2024] [Revised: 08/08/2024] [Accepted: 08/08/2024] [Indexed: 08/21/2024]
Abstract
To control three-dimensional (3D) cell spheroid formation, it is well-known the surface physicochemical and mechanical properties of cell culture materials are important; however, the formation and function of 3D cells are still unclear. This study demonstrated the precise control of the formation of 3D cells and 3D cell functions using diblock copolymers containing different ratios of a zwitterionic trimethylamine N-oxide group. The diblock copolymers were composed of poly(n-butyl methacrylate) (PBMA) as the hydrophobic unit for surface coating on a cell culture dish and stabilization in water, and poly(2-(dimethylamino)ethyl methacrylate) (PDMAEMA) as the precursor of N-oxide. The zwitterionic N-oxide converted from 0 to 100% using PDMAEMA. The wettability and surface zeta potential varied with different ratios of N-oxide diblock copolymer-coated surfaces, and the amount of protein adsorbed in the cell culture medium decreased monotonically with increasing N-oxide ratio. 3D cell spheroid formations were observed by seeding human umbilical cord mesenchymal stem cells (hUC-MSCs) in diblock copolymer-coated flat-bottom well plates, and the N-oxide ratio was over 40%. The cells proliferated in two-dimensions (2D) and did not form spheroids when the N-oxide ratio was less than 20%. Interestingly, the expression of undifferentiated markers of hUC-MSCs was higher on surfaces that adsorbed proteins to some extent and formed 50-150 μm spheroids in the range of 40-70% of N-oxide ratio. We revealed that a moderately protein-adsorbed surface allows precise control of spheroid formation and undifferentiated 3D cells and has potential applications for high-quality spheroids in regenerative medicine and drug screening.
Collapse
Affiliation(s)
- Naoto Ogiwara
- Biotech
Business Unit, Incubation Center, artience
Co., Ltd., 5-6-7 Chiyoda, Sakado-city, Saitama 350-0214, Japan
- Department
of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Takenobu Nakano
- Biotech
Business Unit, Incubation Center, artience
Co., Ltd., 5-6-7 Chiyoda, Sakado-city, Saitama 350-0214, Japan
| | - Koki Baba
- Biotech
Business Unit, Incubation Center, artience
Co., Ltd., 5-6-7 Chiyoda, Sakado-city, Saitama 350-0214, Japan
| | - Hidenori Noguchi
- Center
for Green Research on Energy and Environmental Materials and Global
Research Center for Environment and Energy based on Nanomaterials
Science (GREEN), National Institute for
Materials Science (NIMS), Namiki 1-1, Tsukuba, Ibaraki 305-0044, Japan
| | - Tsukuru Masuda
- Department
of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Madoka Takai
- Department
of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
6
|
Zhou Z, Zhang Y, Zeng Y, Yang D, Mo J, Zheng Z, Zhang Y, Xiao P, Zhong X, Yan W. Effects of Nanomaterials on Synthesis and Degradation of the Extracellular Matrix. ACS NANO 2024; 18:7688-7710. [PMID: 38436232 DOI: 10.1021/acsnano.3c09954] [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: 03/05/2024]
Abstract
Extracellular matrix (ECM) remodeling is accompanied by the continuous synthesis and degradation of the ECM components. This dynamic process plays an important role in guiding cell adhesion, migration, proliferation, and differentiation, as well as in tissue development, body repair, and maintenance of homeostasis. Nanomaterials, due to their photoelectric and catalytic properties and special structure, have garnered much attention in biomedical fields for use in processes such as tissue engineering and disease treatment. Nanomaterials can reshape the cell microenvironment by changing the synthesis and degradation of ECM-related proteins, thereby indirectly changing the behavior of the surrounding cells. This review focuses on the regulatory role of nanomaterials in the process of cell synthesis of different ECM-related proteins and extracellular protease. We discuss influencing factors and possible related mechanisms of nanomaterials in ECM remodeling, which may provide different insights into the design and development of nanomaterials for the treatment of ECM disorder-related diseases.
Collapse
Affiliation(s)
- Zhiyan Zhou
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanli Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510260, China
| | - Yuting Zeng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Dehong Yang
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiayao Mo
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ziting Zheng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuxin Zhang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ping Xiao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xincen Zhong
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenjuan Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
7
|
Asadian M, Tomasina C, Onyshchenko Y, Chan KV, Norouzi M, Zonderland J, Camarero-Espinosa S, Morent R, De Geyter N, Moroni L. The role of plasma-induced surface chemistry on polycaprolactone nanofibers to direct chondrogenic differentiation of human mesenchymal stem cells. J Biomed Mater Res A 2024; 112:210-230. [PMID: 37706337 DOI: 10.1002/jbm.a.37607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/12/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023]
Abstract
Bone marrow-derived mesenchymal stromal cells (BMSCs) are extensively being utilized for cartilage regeneration owing to their excellent differentiation potential and availability. However, controlled differentiation of BMSCs towards cartilaginous phenotypes to heal full-thickness cartilage defects remains challenging. This study investigates how different surface properties induced by either coating deposition or biomolecules immobilization onto nanofibers (NFs) could affect BMSCs chondro-inductive behavior. Accordingly, electrospun poly(ε-caprolactone) (PCL) NFs were exposed to two surface modification strategies based on medium-pressure plasma technology. The first strategy is plasma polymerization, in which cyclopropylamine (CPA) or acrylic acid (AcAc) monomers were plasma polymerized to obtain amine- or carboxylic acid-rich NFs, respectively. The second strategy uses a combination of CPA plasma polymerization and a post-chemical technique to immobilize chondroitin sulfate (CS) onto the NFs. These modifications could affect surface roughness, hydrophilicity, and chemical composition while preserving the NFs' nano-morphology. The results of long-term BMSCs culture in both basic and chondrogenic media proved that the surface modifications modulated BMSCs chondrogenic differentiation. Indeed, the incorporation of polar groups by different modification strategies had a positive impact on the cell proliferation rate, production of the glycosaminoglycan matrix, and expression of extracellular matrix proteins (collagen I and collagen II). The chondro-inductive behavior of the samples was highly dependent on the nature of the introduced polar functional groups. Among all samples, carboxylic acid-rich NFs promoted chondrogenesis by higher expression of aggrecan, Sox9, and collagen II with downregulation of hypertrophic markers. Hence, this approach showed an intrinsic potential to have a non-hypertrophic chondrogenic cell phenotype.
Collapse
Affiliation(s)
- Mahtab Asadian
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
- Prometheus Division of Skeletal Tissue Engineering, Department of Materials Science, KU Leuven University, Leuven, Belgium
| | - Clarissa Tomasina
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
| | - Yuliia Onyshchenko
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Ke Vin Chan
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Mohammad Norouzi
- Department of Pharmacology, University of Montreal, Montreal, Québec, Canada
| | - Jip Zonderland
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
| | - Sandra Camarero-Espinosa
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
- POLYMAT University of the Basque Country UPV/EHU Avenida Tolosa 72, Donostia/San Sebastián, Spain
- IKERBASQUE, Basque Foundation for Science, Euskadi Pl. 5, Bilbao, Spain
| | - Rino Morent
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Nathalie De Geyter
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Lorenzo Moroni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
8
|
Ki MR, Kim SH, Rho S, Kim JK, Min KH, Yeo KB, Lee J, Lee G, Jun SH, Pack SP. Use of biosilica to improve loading and delivery of bone morphogenic protein 2. Int J Biol Macromol 2024; 254:127876. [PMID: 37926322 DOI: 10.1016/j.ijbiomac.2023.127876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
The clinical utility of bone morphogenetic protein 2 (BMP2) is limited because of the poor attraction between BMP2 and carriers, resulting in low loading efficiency and initial burst release. Here, the high binding affinity of BMP2 to the biosilica surface was utilized to overcome this limitation. Atomic force microscopy revealed that BMP2 bound nearly 8- and 2-fold more strongly to biosilica-coated hydroxyapatite than to uncoated and plain silica-coated hydroxyapatite, respectively. To achieve controlled release, collagen was introduced between the silica layers on hydroxyapatite, which was optimized by adjusting the collagen concentration and number of layers. The optimal biosilica/collagen formulation induced sustained BMP2 release without compromising loading efficiency. BMP2 combined with the mentioned formulation led to an increase in osteogenesis, as compared to the combination of BMP2 with either biosilica-coated or non-coated hydroxyapatite in vitro. In rat calvarial defect models, the biosilica/collagen-coated hydroxyapatite with 1 μg BMP2 showed 26 % more bone regeneration than the same dose of BMP2-loaded hydroxyapatite and 10.6 % more than hydroxyapatite with 2.5-fold dose of BMP2. Using BMP2 affinity carriers coated with biosilica/collagen allows for more efficacious in situ loading and delivery of BMP2, making them suitable for the clinical application of growth factors through a soaking method.
Collapse
Affiliation(s)
- Mi-Ran Ki
- Department of Biotechnology and Bioinformatics, Sejong 30019, Republic of Korea; Institute of Industrial Technology, Korea University, Sejong 30019, Republic of Korea
| | - Sung Ho Kim
- Department of Biotechnology and Bioinformatics, Sejong 30019, Republic of Korea
| | - Seokbeom Rho
- Department of Biotechnology and Bioinformatics, Sejong 30019, Republic of Korea; Interdisciplinary Graduate Program for Artificial Intelligence Smart Convergence Technology, Sejong 30019, Republic of Korea
| | - Jong Ki Kim
- Department of Biotechnology and Bioinformatics, Sejong 30019, Republic of Korea
| | - Ki Ha Min
- Department of Biotechnology and Bioinformatics, Sejong 30019, Republic of Korea
| | - Ki Baek Yeo
- Department of Oral and Maxillofacial Surgery, Korea University Anam Hospital, Seoul 02841, Republic of Korea
| | - Jaewook Lee
- Department of Oral and Maxillofacial Surgery, Korea University Anam Hospital, Seoul 02841, Republic of Korea
| | - Gyudo Lee
- Department of Biotechnology and Bioinformatics, Sejong 30019, Republic of Korea; Interdisciplinary Graduate Program for Artificial Intelligence Smart Convergence Technology, Sejong 30019, Republic of Korea
| | - Sang-Ho Jun
- Department of Oral and Maxillofacial Surgery, Korea University Anam Hospital, Seoul 02841, Republic of Korea.
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Sejong 30019, Republic of Korea.
| |
Collapse
|
9
|
Zhao Q, Zhao Z, Zhang J, Ni Y, Ouyang S, Qi H, Yu Y, Miron RJ, Tang H, Zhang Y. Fn-HMGB1 Adsorption Behavior Initiates Early Immune Recognition and Subsequent Osteoinduction of Biomaterials. Adv Healthc Mater 2024; 13:e2301808. [PMID: 37602504 DOI: 10.1002/adhm.202301808] [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: 06/07/2023] [Revised: 08/11/2023] [Indexed: 08/22/2023]
Abstract
Implantable biomaterials are widely used in bone tissue engineering, but little is still known about how they initiate early immune recognition and the initial dynamics. Herein, the early immune recognition and subsequent osteoinduction of biphasic calcium phosphate (BCP) after implantation to the protein adsorption behavior is attributed. By liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis, the biomaterial-related molecular patterns (BAMPs) formed after BCP implantation are mapped, dominated by the highly expressed extracellular matrix protein fibronectin (Fn) and the high mobility group box 1 (HMGB1). Molecular dynamics simulations show that Fn has the ability to bind more readily to the BCP surface than HMGB1. The preferential binding of Fn provides a higher adsorption energy for HMGB1. Furthermore, multiple hydrogen bonding sites between HMGB1 and Fn are demonstrated using a molecular docking approach. Ultimately, the formation of BAMPs through HMGB1 antagonist glycyrrhizic acid (GA), resulting in impaired immune recognition of myeloid differentiation factor 88 (MYD88) mediated dendritic cells (DCs) and macrophages (Mφs), as well as failed osteoinduction processes is obstructed. This study introduces a mechanism for early immune recognition of implant materials based on protein adsorption, providing perspectives for future design and application of tissue engineering materials.
Collapse
Affiliation(s)
- 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
| | - Zifan Zhao
- Center of Digital Dentistry, Faculty of Prosthodontics, Peking University School and Hospital of Stomatology; National Center of Stomatology; National Engineerœing 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, National Clinical Research Center for Oral Diseases, Beijing, 100081, China
| | - Jing 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
| | - Yueqi Ni
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Simin Ouyang
- 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
| | - Haoning Qi
- 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
| | - Yiqian Yu
- 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
| | - Richard J Miron
- Department of Periodontology, University of Bern, Bern, 300392, Switzerland
| | - Hua Tang
- Department of Rheumatology and Autoimmunology, Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250000, China
- Institute of Infection and Immunity, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, 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
| |
Collapse
|
10
|
Qiao Y, Yu L, Yang P, Chen M, Sun H, Wang L, Wu B, Oh C, Yang H, Bai J, Geng D. Spatiotemporal Immunomodulation and Biphasic Osteo-Vascular Aligned Electrospun Membrane for Diabetic Periosteum Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302874. [PMID: 37973554 PMCID: PMC10754081 DOI: 10.1002/advs.202302874] [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: 05/08/2023] [Revised: 07/10/2023] [Indexed: 11/19/2023]
Abstract
Under diabetic conditions, blood glucose fluctuations and exacerbated immunopathological inflammatory environments pose significant challenges to periosteal regenerative repair strategies. Responsive immune regulation in damaged tissues is critical for the immune microenvironment, osteogenesis, and angiogenesis stabilization. Considering the high-glucose microenvironment of such acute injury sites, a functional glucose-responsive immunomodulation-assisted periosteal regeneration composite material-PLA(Polylactic Acid)/COLI(Collagen I)/Lipo(Liposome)-APY29 (PCLA)-is constructed. Aside from stimulating osteogenic differentiation, owing to the presence of surface self-assembled type I collagen in the scaffolds, PCLA can directly respond to focal area high-glucose microenvironments. The PCLA scaffolds trigger the release of APY29-loaded liposomes, shifting the macrophages toward the M2 phenotype, inhibiting the release of inflammatory cytokines, improving the bone immune microenvironment, and promoting osteogenic differentiation and angiogenesis. Bioinformatics analyses show that PCLA enhances bone repair by inhibiting the inflammatory signal pathway regulating the polarization direction and promoting osteogenic and angiogenic gene expression. In the calvarial periosteal defect model of diabetic rats, PCLA scaffolds induce M2 macrophage polarization and improve the inflammatory microenvironment, significantly accelerating periosteal repair. Overall, the PCLA scaffold material regulates immunity in fluctuating high-glucose inflammatory microenvironments, achieves relatively stable and favorable osteogenic microenvironments, and facilitates the effective design of functionalized biomaterials for bone regeneration therapy in patients with diabetes.
Collapse
Affiliation(s)
- Yusen Qiao
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
- Department of Orthopedic SurgeryRush University Medical CenterChicagoIL60612USA
| | - Lei Yu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Peng Yang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Miao Chen
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Haifu Sun
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Lingjie Wang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Bangzhao Wu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Chun‐do Oh
- Department of Orthopedic SurgeryRush University Medical CenterChicagoIL60612USA
| | - Huilin Yang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| | - Jiaxiang Bai
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230022China
- National Center for Translational Medicine (Shanghai) SHU BranchShanghai UniversityShanghaiChina
| | - Dechun Geng
- Department of OrthopedicsThe First Affiliated Hospital of Soochow University188 Shizi RoadSuzhouJiangsu215006China
| |
Collapse
|
11
|
Tripathi AS, Zaki MEA, Al-Hussain SA, Dubey BK, Singh P, Rind L, Yadav RK. Material matters: exploring the interplay between natural biomaterials and host immune system. Front Immunol 2023; 14:1269960. [PMID: 37936689 PMCID: PMC10627157 DOI: 10.3389/fimmu.2023.1269960] [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: 07/31/2023] [Accepted: 10/02/2023] [Indexed: 11/09/2023] Open
Abstract
Biomaterials are widely used for various medical purposes, for instance, implants, tissue engineering, medical devices, and drug delivery systems. Natural biomaterials can be obtained from proteins, carbohydrates, and cell-specific sources. However, when these biomaterials are introduced into the body, they trigger an immune response which may lead to rejection and failure of the implanted device or tissue. The immune system recognizes natural biomaterials as foreign substances and triggers the activation of several immune cells, for instance, macrophages, dendritic cells, and T cells. These cells release pro-inflammatory cytokines and chemokines, which recruit other immune cells to the implantation site. The activation of the immune system can lead to an inflammatory response, which can be beneficial or detrimental, depending on the type of natural biomaterial and the extent of the immune response. These biomaterials can also influence the immune response by modulating the behavior of immune cells. For example, biomaterials with specific surface properties, such as charge and hydrophobicity, can affect the activation and differentiation of immune cells. Additionally, biomaterials can be engineered to release immunomodulatory factors, such as anti-inflammatory cytokines, to promote a tolerogenic immune response. In conclusion, the interaction between biomaterials and the body's immune system is an intricate procedure with potential consequences for the effectiveness of therapeutics and medical devices. A better understanding of this interplay can help to design biomaterials that promote favorable immune responses and minimize adverse reactions.
Collapse
Affiliation(s)
| | - Magdi E A Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| | - Sami A Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad lbn Saud Islamic University, Riyadh, Saudi Arabia
| | - Bidhyut Kumar Dubey
- Department of Pharmaceutical Chemistry, Era College of Pharmacy, Era University, Lucknow, India
| | - Prabhjot Singh
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, India
| | - Laiba Rind
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, India
| | - Rajnish Kumar Yadav
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, India
| |
Collapse
|
12
|
Hanetseder D, Levstek T, Teuschl-Woller AH, Frank JK, Schaedl B, Redl H, Marolt Presen D. Engineering of extracellular matrix from human iPSC-mesenchymal progenitors to enhance osteogenic capacity of human bone marrow stromal cells independent of their age. Front Bioeng Biotechnol 2023; 11:1214019. [PMID: 37600321 PMCID: PMC10434254 DOI: 10.3389/fbioe.2023.1214019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Regeneration of bone defects is often limited due to compromised bone tissue physiology. Previous studies suggest that engineered extracellular matrices enhance the regenerative capacity of mesenchymal stromal cells. In this study, we used human-induced pluripotent stem cells, a scalable source of young mesenchymal progenitors (hiPSC-MPs), to generate extracellular matrix (iECM) and test its effects on the osteogenic capacity of human bone-marrow mesenchymal stromal cells (BMSCs). iECM was deposited as a layer on cell culture dishes and into three-dimensional (3D) silk-based spongy scaffolds. After decellularization, iECM maintained inherent structural proteins including collagens, fibronectin and laminin, and contained minimal residual DNA. Young adult and aged BMSCs cultured on the iECM layer in osteogenic medium exhibited a significant increase in proliferation, osteogenic marker expression, and mineralization as compared to tissue culture plastic. With BMSCs from aged donors, matrix mineralization was only detected when cultured on iECM, but not on tissue culture plastic. When cultured in 3D iECM/silk scaffolds, BMSCs exhibited significantly increased osteogenic gene expression levels and bone matrix deposition. iECM layer showed a similar enhancement of aged BMSC proliferation, osteogenic gene expression, and mineralization compared with extracellular matrix layers derived from young adult or aged BMSCs. However, iECM increased osteogenic differentiation and decreased adipocyte formation compared with single protein substrates including collagen and fibronectin. Together, our data suggest that the microenvironment comprised of iECM can enhance the osteogenic activity of BMSCs, providing a bioactive and scalable biomaterial strategy for enhancing bone regeneration in patients with delayed or failed bone healing.
Collapse
Affiliation(s)
- Dominik Hanetseder
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Tina Levstek
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andreas Herbert Teuschl-Woller
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
| | - Julia Katharina Frank
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Barbara Schaedl
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Darja Marolt Presen
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
13
|
Wang K, Man K, Liu J, Meckes B, Yang Y. Dissecting Physical and Biochemical Effects in Nanotopographical Regulation of Cell Behavior. ACS NANO 2023; 17:2124-2133. [PMID: 36668987 DOI: 10.1021/acsnano.2c08075] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Regulating cell behavior using nanotopography has been widely implemented. To facilitate cell adhesion, physical nanotopography is usually coated with adhesive proteins such as fibronectin (FN). However, the confounding effects of physical and biochemical cues of nanotopography hinder the understanding of nanotopography in regulating cell behavior, which ultimately limits the biomedical applications of nanotopography. To delineate the roles of the physical and biochemical cues in cell regulation, we fabricate substrates that have either the same physical nanotopography but different biochemical (FN) nanopatterns or identical FN nanopatterns but different physical nanotopographies. We then examine the influences of physical and biochemical cues of nanotopography on spreading, nuclear deformation, mechanotransduction, and function of human mesenchymal stem cells (hMSCs). Our results reveal that physical topographies, especially nanogratings, dominantly control cell spreading, YAP localization, proliferation, and differentiation of hMSCs. However, biochemical FN nanopatterns affect hMSC elongation, YAP intracellular localization, and lamin a/c (LAMAC) expression. Furthermore, we find that physical nanogratings induce nanoscale curvature of nuclei at the basal side, which attenuates the osteogenic differentiation of hMSCs. Collectively, our study highlights the dominant effect of physical nanotopography in regulating stem cell functions, while suggesting that fine-tuning of cell behavior can be achieved through altering the presentation of biochemical cues on substrate surfaces.
Collapse
Affiliation(s)
- Kai Wang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Brian Meckes
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
14
|
de Souza W, Gemini-Piperni S, Grenho L, Rocha LA, Granjeiro JM, Melo SA, Fernandes MH, Ribeiro AR. Titanium dioxide nanoparticles affect osteoblast-derived exosome cargos and impair osteogenic differentiation of human mesenchymal stem cells. Biomater Sci 2023; 11:2427-2444. [PMID: 36756939 DOI: 10.1039/d2bm01854c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Titanium (Ti) and its alloys are the most widely used metallic biomaterials in total joint replacement; however, increasing evidence supports the degradation of its surface due to corrosion and wear processes releasing debris (ions, and micro and nanoparticles) and contribute to particle-induced osteolysis and implant loosening. Cell-to-cell communication involving several cell types is one of the major biological processes occurring during bone healing and regeneration at the implant-bone interface. In addition to the internal response of cells to the uptake and intracellular localization of wear debris, a red flag is the ability of titanium dioxide nanoparticles (mimicking wear debris) to alter cellular communication with the tissue background, disturbing the balance between osseous tissue integrity and bone regenerative processes. This study aims to understand whether titanium dioxide nanoparticles (TiO2 NPs) alter osteoblast-derived exosome (Exo) biogenesis and whether exosomal protein cargos affect the communication of osteoblasts with human mesenchymal stem/stromal cells (HMSCs). Osteoblasts are derived from mesenchymal stem cells coexisting in the bone microenvironment during development and remodelling. We observed that TiO2 NPs stimulate immature osteoblast- and mature osteoblast-derived Exo secretion that present a distinct proteomic cargo. Functional tests confirmed that Exos derived from both osteoblasts decrease the osteogenic differentiation of HMSCs. These findings are clinically relevant since wear debris alter extracellular communication in the bone periprosthetic niche, contributing to particle-induced osteolysis and consequent prosthetic joint failure.
Collapse
Affiliation(s)
- Wanderson de Souza
- Directory of Metrology Applied to Life Sciences, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil.,Postgraduate Program in Biotechnology, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil
| | - S Gemini-Piperni
- Postgraduate Program in Biotechnology, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil.,Postgraduate Program in Translational Biomedicine, University Grande Rio, Duque de Caxias, Brazil.,Lab∈n Group, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil
| | - Liliana Grenho
- Faculty of Dental Medicine, University of Porto, Porto, Portugal.,LAQV/REQUIMTE, University of Porto, Porto, Portugal
| | - Luís A Rocha
- Physics Department, Paulista State University, São Paulo, Brazil.,IBTN/Br - Brazilian Branch of the Institute of Biomaterials, Tribocorrosion and Nanomedicine, São Paulo State University, Bauru, São Paulo, Brazil
| | - José M Granjeiro
- Directory of Metrology Applied to Life Sciences, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil.,Postgraduate Program in Biotechnology, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil.,Postgraduate Program in Translational Biomedicine, University Grande Rio, Duque de Caxias, Brazil.,Dental School, Fluminense Federal University, Niterói, Brazil
| | - Sonia A Melo
- i3S-Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Maria H Fernandes
- Faculty of Dental Medicine, University of Porto, Porto, Portugal.,LAQV/REQUIMTE, University of Porto, Porto, Portugal
| | - Ana R Ribeiro
- Postgraduate Program in Biotechnology, National Institute of Metrology Quality and Technology, Rio de Janeiro, Brazil.,NanoSafety group, International Iberian Nanotechnology Laboratory - INL, 4715-330, Braga, Portugal.
| |
Collapse
|
15
|
Hu J, Liu S, Fan C. Applications of functionally-adapted hydrogels in tendon repair. Front Bioeng Biotechnol 2023; 11:1135090. [PMID: 36815891 PMCID: PMC9934866 DOI: 10.3389/fbioe.2023.1135090] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Despite all the efforts made in tissue engineering for tendon repair, the management of tendon injuries still poses a challenge, as current treatments are unable to restore the function of tendons following injuries. Hydrogels, due to their exceptional biocompatibility and plasticity, have been extensively applied and regarded as promising candidate biomaterials in tissue regeneration. Varieties of approaches have designed functionally-adapted hydrogels and combined hydrogels with other factors (e.g., bioactive molecules or drugs) or materials for the enhancement of tendon repair. This review first summarized the current state of knowledge on the mechanisms underlying the process of tendon healing. Afterward, we discussed novel strategies in fabricating hydrogels to overcome the issues frequently encountered during the applications in tendon repair, including poor mechanical properties and undesirable degradation. In addition, we comprehensively summarized the rational design of hydrogels for promoting stem-cell-based tendon tissue engineering via altering biophysical and biochemical factors. Finally, the role of macrophages in tendon repair and how they respond to immunomodulatory hydrogels were highlighted.
Collapse
Affiliation(s)
- Jiacheng Hu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Shen Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| |
Collapse
|
16
|
Zhao H, Wang X, Jin A, Wang M, Wang Z, Huang X, Dai J, Wang X, Lin D, Shen SGF. Reducing relapse and accelerating osteogenesis in rapid maxillary expansion using an injectable mesoporous bioactive glass/fibrin glue composite hydrogel. Bioact Mater 2022; 18:507-525. [PMID: 35415307 PMCID: PMC8976096 DOI: 10.1016/j.bioactmat.2022.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 11/29/2022] Open
Abstract
Rapid maxillary expansion (RME), as a common treatment for craniomaxillofacial deformity, faces the challenge of high relapse rates and unsatisfactory therapeutic effects. In this study, a standardized Sprague-Dawley (SD) rat RME model was first established with a modified expander as well as retainer design and optimized anterior maxillary expanding force of 100 g which exerted the most synchronized mobility of mid-palatal suture and incisors. Via the standardized model, the high relapse rate was proven to be attributed to insufficient osteogenesis in expanded suture, requiring long-term retainer wearing in clinical situations. To reduce the relapse rate, mesoporous bioactive glass/fibrin glue (MBG/FG) composite hydrogels were developed for an in situ minimal invasive injection that enhance osteogenesis in the expanded palate. The component of 1 wt% MBG was adopted for enhanced mechanical strength, matched degradation rate and ion dissolution, excellent in vitro biocompatibility and osteoinductivity. Effects of 1%MBG/FG composite hydrogel on osteogenesis in expanded mid-palatal sutures with/without retention were evaluated in the standardized model. The results demonstrated that injection of 1%MBG/FG composite hydrogel significantly promoted bone formation within the expanded mid-palatal suture, inhibited osteoclastogenesis and benefited the balance of bone remodeling towards osteogenesis. Combination of retainer and injectable biomaterial was demonstrated as a promising treatment to reduce relapse rate and enhance osteogenesis after RME. The model establishment and the composite hydrogel development in this article might provide new insight to other craniomaxillofacial deformity treatment and design of bone-repairing biomaterials with higher regenerative efficiency. A standardized rat RME model was established with optimized parameters. Sufficient osteogenesis was the prerequisite of reducing relapse ratio. Design of an injectable MBG/FG composite hydrogel for osteogenic enhancement. Combinatory treatment of injection and retention was developed for relapse reduction.
Collapse
|
17
|
Abbasi-Ravasjani S, Seddiqi H, Moghaddaszadeh A, Ghiasvand ME, Jin J, Oliaei E, Bacabac RG, Klein-Nulend J. Sulfated carboxymethyl cellulose and carboxymethyl κ-carrageenan immobilization on 3D-printed poly-ε-caprolactone scaffolds differentially promote pre-osteoblast proliferation and osteogenic activity. Front Bioeng Biotechnol 2022; 10:957263. [PMID: 36213076 PMCID: PMC9542643 DOI: 10.3389/fbioe.2022.957263] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
The lack of bioactivity in three-dimensional (3D)-printing of poly-є-caprolactone (PCL) scaffolds limits cell-material interactions in bone tissue engineering. This constraint can be overcome by surface-functionalization using glycosaminoglycan-like anionic polysaccharides, e.g., carboxymethyl cellulose (CMC), a plant-based carboxymethylated, unsulfated polysaccharide, and κ-carrageenan, a seaweed-derived sulfated, non-carboxymethylated polysaccharide. The sulfation of CMC and carboxymethylation of κ-carrageenan critically improve their bioactivity. However, whether sulfated carboxymethyl cellulose (SCMC) and carboxymethyl κ-carrageenan (CM-κ-Car) affect the osteogenic differentiation potential of pre-osteoblasts on 3D-scaffolds is still unknown. Here, we aimed to assess the effects of surface-functionalization by SCMC or CM-κ-Car on the physicochemical and mechanical properties of 3D-printed PCL scaffolds, as well as the osteogenic response of pre-osteoblasts. MC3T3-E1 pre-osteoblasts were seeded on 3D-printed PCL scaffolds that were functionalized by CM-κ-Car (PCL/CM-κ-Car) or SCMC (PCL/SCMC), cultured up to 28 days. The scaffolds’ physicochemical and mechanical properties and pre-osteoblast function were assessed experimentally and by finite element (FE) modeling. We found that the surface-functionalization by SCMC and CM-κ-Car did not change the scaffold geometry and structure but decreased the elastic modulus. Furthermore, the scaffold surface roughness and hardness increased and the scaffold became more hydrophilic. The FE modeling results implied resilience up to 2% compression strain, which was below the yield stress for all scaffolds. Surface-functionalization by SCMC decreased Runx2 and Dmp1 expression, while surface-functionalization by CM-κ-Car increased Cox2 expression at day 1. Surface-functionalization by SCMC most strongly enhanced pre-osteoblast proliferation and collagen production, while CM-κ-Car most significantly increased alkaline phosphatase activity and mineralization after 28 days. In conclusion, surface-functionalization by SCMC or CM-κ-Car of 3D-printed PCL-scaffolds enhanced pre-osteoblast proliferation and osteogenic activity, likely due to increased surface roughness and hydrophilicity. Surface-functionalization by SCMC most strongly enhanced cell proliferation, while CM-κ-Car most significantly promoted osteogenic activity, suggesting that surface-functionalization by CM-κ-Car may be more promising, especially in the short-term, for in vivo bone formation.
Collapse
Affiliation(s)
- Sonia Abbasi-Ravasjani
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam Movement Sciences, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Hadi Seddiqi
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam Movement Sciences, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ali Moghaddaszadeh
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Jianfeng Jin
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam Movement Sciences, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Erfan Oliaei
- Fiber and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Rommel Gaud Bacabac
- Medical Biophysics Group, Department of Physics, University of San Carlos, Cebu City, Philippines
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam Movement Sciences, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- *Correspondence: Jenneke Klein-Nulend,
| |
Collapse
|
18
|
Lim R, Banerjee A, Biswas R, Chari AN, Raghavan S. Mechanotransduction through adhesion molecules: Emerging roles in regulating the stem cell niche. Front Cell Dev Biol 2022; 10:966662. [PMID: 36172276 PMCID: PMC9511051 DOI: 10.3389/fcell.2022.966662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Stem cells have been shown to play an important role in regenerative medicine due to their proliferative and differentiation potential. The challenge, however, lies in regulating and controlling their potential for this purpose. Stem cells are regulated by growth factors as well as an array of biochemical and mechanical signals. While the role of biochemical signals and growth factors in regulating stem cell homeostasis is well explored, the role of mechanical signals has only just started to be investigated. Stem cells interact with their niche or to other stem cells via adhesion molecules that eventually transduce mechanical cues to maintain their homeostatic function. Here, we present a comprehensive review on our current understanding of the influence of the forces perceived by cell adhesion molecules on the regulation of stem cells. Additionally, we provide insights on how this deeper understanding of mechanobiology of stem cells has translated toward therapeutics.
Collapse
Affiliation(s)
- Ryan Lim
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Avinanda Banerjee
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Ritusree Biswas
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
- Sastra University, Thanjavur, TN, India
| | - Anana Nandakumar Chari
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Srikala Raghavan
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
| |
Collapse
|
19
|
Yang Y, Chen D, Li Y, Zou J, Han R, Li H, Zhang J. Effect of Puerarin on Osteogenic Differentiation in vitro and on New Bone Formation in vivo. Drug Des Devel Ther 2022; 16:2885-2900. [PMID: 36060929 PMCID: PMC9433167 DOI: 10.2147/dddt.s379794] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/17/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose Puerarin (C21H20O10) is a phytoestrogen that possesses various pharmacological effect, and several researches have revealed the relationship between puerarin and bone metabolism. This study was aimed to evaluate the potential influence of puerarin on the proliferation and osteogenic differentiation of rat bone marrow-derived mesenchymal stem cells (BMSCs) as well as on new bone formation following rapid maxillary expansion (RME) model in rats. Methods Rat BMSCs were adopted, and the cell proliferation was detected by cell-counting kit-8 (CCK-8) assay in vitro experiments. Alkaline phosphatase (ALP) activity and alizarin red staining were analyzed quantitatively to show extracellular matrix mineralization. The mRNA and protein expression levels were used to detect osteogenic differentiation of BMSCs. In vivo bone regeneration was analyzed in a rat RME model. Eighteen 6-week-old male Wistar rats were divided into 3 groups: group 1 without any treatment, group 2 received RME and saline solution (15mg/kg), group 3 received RME and puerarin solution (15mg/kg). After 2 weeks, micro-computed tomography (Micro-CT), hematoxylin and eosin (HE) staining, and Masson staining were used to detect the new bone formation and morphological changes. Besides, ALP and bone morphogenetic protein 2 (BMP2) expression levels in mid-palatal suture were evaluated by immunohistochemical staining. Results The results showed that puerarin upregulates cell proliferation dose-dependently. ALP activity and mineralized matrix generation were clearly enhanced at certain specific concentrations (10−5 and 10−6 mol/L); the expression levels of the osteoblast-related genes and proteins were increased. The measurement of micro-CT imaging revealed that puerarin significantly promoted new bone formation. Concomitantly, the histological examinations showed that puerarin solution enhanced osteogenesis in mid-palatal suture. Conclusion Those works indicated that puerarin regulates osteogenesis in vitro and exerts a beneficial impact on bone regeneration in vivo, revealing that puerarin treatment may become one of the potential keys for improving the stability and preventing relapse of RME.
Collapse
Affiliation(s)
- Yanran Yang
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Daiyun Chen
- Department of Orthodontics, School of Stomatology, Shandong First Medical University, Jinan, People’s Republic of China
| | - Yilin Li
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Jinghua Zou
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Ruiqi Han
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Hongkun Li
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Jun Zhang
- Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
- Correspondence: Jun Zhang, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China, Tel +86 13953109816, Fax +86 53188382923, Email
| |
Collapse
|
20
|
Bang JY, Youn KE, Kim RH, Song M. Effect of the amnion-chorion or collagen membrane as a matrix on the microenvironment during a regenerative endodontic procedure. J Endod 2022; 48:1285-1293.e2. [PMID: 35850299 DOI: 10.1016/j.joen.2022.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 12/29/2022]
Abstract
INTRODUCTION During regenerative endodontic procedures, the microenvironment of the canal is formed by the degree of disinfection and release of ions from the applied materials onto the top surface. This study aimed to characterize the effects of amnion-chorion membrane (ACM) and collagen membrane (CM) on pulp-dentin regeneration compared to calcium silicate cements (CSCs), focusing on cell migration, mineralization potential, anti-inflammation, and angiogenesis. METHODS Two CSCs and two membranes were used: ProRoot® MTA (PM; Dentsply, Tulsa, OK, USA), RetroMTA® (RM; BioMTA, Seoul, Korea), Collagen Membrane® (CM; Genoss, Suwon, Korea), and BioXclude® (ACM; Snoasis Medical, Colorado, USA). Transwell and scratch assays were used to evaluate cell migration and wound healing. Mineralization potential was evaluated using alkaline phosphatase (ALP) activity, alizarin red S staining, and quantitative real-time-polymerase chain reaction (qRT-PCR) for the expression of marker genes. qRT-PCR was used to measure the levels of angiogenic genes and inflammatory mediators. An endothelial tube formation assay was used to assess angiogenesis. RESULTS The membranes showed superior migration and wound healing compared with CSCs. Except for RM, PM and the two membranes showed high ALP activity and mineralization nodule formation and upregulated mRNA expression of markers for mineralization. Membranes upregulated the mRNA of angiogenesis genes and increased the capillary tube formation of endothelial cells compared to CSCs. Furthermore, the membrane matrix decreased the expression of inflammatory genes. CONCLUSIONS CM and ACM showed prominent cell migration, angiogenesis, and healing effects against inflammation, as well as comparable mineralization potential compared to CSCs, recommending the use of membrane as a matrix.
Collapse
Affiliation(s)
- Ji Young Bang
- Department of Conservative Dentistry, College of Dentistry, Dankook University, Cheonan, Korea
| | - Kyeong Eun Youn
- Department of Conservative Dentistry, College of Dentistry, Dankook University, Cheonan, Korea
| | - Reuben H Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA, USA; Section of Restorative Dentistry, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Minju Song
- Department of Conservative Dentistry, College of Dentistry, Dankook University, Cheonan, Korea; Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Korea.
| |
Collapse
|
21
|
Maalouf M, Abou Khalil A, Di Maio Y, Papa S, Sedao X, Dalix E, Peyroche S, Guignandon A, Dumas V. Polarization of Femtosecond Laser for Titanium Alloy Nanopatterning Influences Osteoblastic Differentiation. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1619. [PMID: 35630841 PMCID: PMC9147489 DOI: 10.3390/nano12101619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 12/23/2022]
Abstract
Ultrashort pulse lasers have significant advantages over conventional continuous wave and long pulse lasers for the texturing of metallic surfaces, especially for nanoscale surface structure patterning. Furthermore, ultrafast laser beam polarization allows for the precise control of the spatial alignment of nanotextures imprinted on titanium-based implant surfaces. In this article, we report the biological effect of beam polarization on human mesenchymal stem cell differentiation. We created, on polished titanium-6aluminum-4vanadium (Ti-6Al-4V) plates, a laser-induced periodic surface structure (LIPSS) using linear or azimuthal polarization of infrared beams to generate linear or radial LIPSS, respectively. The main difference between the two surfaces was the microstructural anisotropy of the linear LIPSS and the isotropy of the radial LIPSS. At 7 d post seeding, cells on the radial LIPSS surface showed the highest extracellular fibronectin production. At 14 days, qRT-PCR showed on the same surface an increase in osteogenesis-related genes, such as alkaline phosphatase and osterix. At 21 d, mineralization clusters indicative of final osteoinduction were more abundant on the radial LIPSS. Taken together, we identified that creating more isotropic than linear surfaces enhances cell differentiation, resulting in an improved osseointegration. Thus, the fine tuning of ultrashort pulse lasers may be a promising new route for the functionalization of medical implants.
Collapse
Affiliation(s)
- Mathieu Maalouf
- SAINBIOSE Laboratory INSERM U1509, Jean Monnet University, University of Lyon, F-42270 Saint Priest en Jarez, France; (S.P.); (E.D.); (S.P.); (A.G.)
| | - Alain Abou Khalil
- Hubert-Curien Laboratory, Jean Monnet University, University of Lyon, UMR 5516 CNRS, F-42000 Saint-Etienne, France; (A.A.K.); (X.S.)
| | - Yoan Di Maio
- GIE Manutech-USD, F-42000 Saint-Etienne, France;
| | - Steve Papa
- SAINBIOSE Laboratory INSERM U1509, Jean Monnet University, University of Lyon, F-42270 Saint Priest en Jarez, France; (S.P.); (E.D.); (S.P.); (A.G.)
| | - Xxx Sedao
- Hubert-Curien Laboratory, Jean Monnet University, University of Lyon, UMR 5516 CNRS, F-42000 Saint-Etienne, France; (A.A.K.); (X.S.)
- GIE Manutech-USD, F-42000 Saint-Etienne, France;
| | - Elisa Dalix
- SAINBIOSE Laboratory INSERM U1509, Jean Monnet University, University of Lyon, F-42270 Saint Priest en Jarez, France; (S.P.); (E.D.); (S.P.); (A.G.)
| | - Sylvie Peyroche
- SAINBIOSE Laboratory INSERM U1509, Jean Monnet University, University of Lyon, F-42270 Saint Priest en Jarez, France; (S.P.); (E.D.); (S.P.); (A.G.)
| | - Alain Guignandon
- SAINBIOSE Laboratory INSERM U1509, Jean Monnet University, University of Lyon, F-42270 Saint Priest en Jarez, France; (S.P.); (E.D.); (S.P.); (A.G.)
| | - Virginie Dumas
- Laboratory of Tribology and Systems Dynamics, Ecole Nationale d’Ingénieurs de Saint Etienne, Ecole Centrale de Lyon, University of Lyon, UMR 5513 CNRS, F-42100 Saint-Etienne, France;
| |
Collapse
|
22
|
Niu Q, He J, Wu M, Liu J, Lu X, Zhang L, Jin Z. Transplantation of bone marrow mesenchymal stem cells and fibrin glue into extraction socket in maxilla promoted bone regeneration in osteoporosis rat. Life Sci 2022; 290:119480. [PMID: 33862113 DOI: 10.1016/j.lfs.2021.119480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 03/03/2021] [Accepted: 03/31/2021] [Indexed: 11/20/2022]
Abstract
AIMS Bone defect repair in osteoporosis remains a tremendous challenge for clinicians due to increased bone metabolism resulted from estrogen deficiency. This study aims to investigate the effect of bone marrow mesenchymal stem cells (BMSCs) combined with fibrin glue (FG) in the extraction socket healing process of osteoporosis rats, as well as estimate the role of estrogen receptors (ERs) played in BMSCs differentiation in vitro and in the alveolar bone reconstruction process in vivo. MAIN METHODS Forty rats were randomly divided into four groups, under general anesthesia, three groups underwent bilateral ovariectomy(OVX) and one group with the sham operation. Three months later, the osteogenic ability of BMSCs, isolated from healthy and osteoporosis rats, respectively, was tested. The ERα and ERβ mRNA expression in BMSCs was also evaluated by RT-PCR analysis. In vivo experiment, Micro-CT detection, histological and immunofluorescent analysis, tissue PCR was conducted up to 2, 4 and 6 weeks after transplantation of BMSCs/FG to assess the newly formed bone in the extraction socket. KEY FINDINGS The BMSCs from osteoporosis rats displayed weaker osteogenic potential and lower ERs expression compared with the BMSCs from healthy rats. Newly formed bone tissue filled the socket defect in BMSCs/FG treated VOX rats after six weeks, which was comparable to the sham group, while reduced ERs expression was found in the regenerated bone of the OVX group. SIGNIFICANCE The BMSCs seeded within FG might provide an alternative therapeutic method for repairing the extraction socket defect in osteoporosis condition.
Collapse
Affiliation(s)
- Qiannan Niu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jiaojiao He
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Minke Wu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Pediatric Dentistry, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jia Liu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xiaolin Lu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Liang Zhang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Stomatology, The Air Force 986 Hospital, Xi'an, People's Republic of China.
| | - Zuolin Jin
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China.
| |
Collapse
|
23
|
Making Sense of the Highly Variable Effects of Alcohol on Bone. Clin Rev Bone Miner Metab 2021. [DOI: 10.1007/s12018-021-09277-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
24
|
Collagen type I promotes osteogenic differentiation of amniotic membrane-derived mesenchymal stromal cells in basal and induction media. Biosci Rep 2021; 40:227060. [PMID: 33245097 PMCID: PMC7736623 DOI: 10.1042/bsr20201325] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
Collagen has been widely shown to promote osteogenesis of bone marrow mesenchymal stromal cells (BM-MSCs). Due to the invasive procedure of obtaining BM-MSCs, MSCs from other tissues have emerged as a promising alternative for regenerative therapy. MSCs originated from different sources, exhibiting different differentiation potentials. Therefore, the applicability of collagen type I (COL), combining with amniotic membrane (AM)-MSCs was examined through proliferation and differentiation assays together with the expression of surface markers and genes associated with stemness and differentiation under basal or induction conditions. No increase in cell growth was observed because AM-MSCs might be directed toward spontaneous osteogenesis. This was evidenced by the calcium deposition and elevated expression of osteogenic genes when AM-MSCs were cultured in collagen plate with basal media. Under the osteogenic condition, reciprocal expression of OCN and CEBPA suggested a shift toward adipogenesis. Surprisingly, adipogenic genes were not elevated upon adipogenic induction, although oil droplets deposition was observed. In conclusion, our findings demonstrated that collagen causes spontaneous osteogenesis in AM-MSCs. However, the presence of exogenous inductors could shift the direction of adipo-osteogenic gene regulatory network modulated by collagen.
Collapse
|
25
|
Expression of CD146 and Regenerative Cytokines by Human Placenta-Derived Mesenchymal Stromal Cells upon Expansion in Different GMP-Compliant Media. Stem Cells Int 2021; 2021:6662201. [PMID: 33868409 PMCID: PMC8035028 DOI: 10.1155/2021/6662201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/08/2021] [Accepted: 03/17/2021] [Indexed: 11/18/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been successfully employed in clinical applications. In most studies, autologous MSCs from the bone marrow (bmMSCs) were used, and others employed autologous adipose tissue-derived stromal cells (ADSCs). Recently, clinical feasibility studies provided evidence that MSCs from human term placenta (pMSCs) can be used for homologous therapy facilitating access to regenerative cells in emergency situations, when autologous cells are not available or not suitable. We therefore investigated the expression of MSC stemness marker CD146 and the expression of neuro- and myoregenerative cytokines by human pMSCs after expansion in three different media compliant with good manufacturing protocols (GMP) in comparison to pMSCs expanded in a commercial MSC expansion media. To replace xenobiotic serum in the GMP-compliant media employed in this study, either human serum, human serum plus platelet lysate (PLL), or human plasma plus PLL was used. We report that enrichment of media with PLL accelerates pMSC proliferation but reduces the expression of the stemness marker CD146 significantly, while PLL deprivation enhanced the CD146 expression. In contrast, the reduced expression of CD146 by PLL deprivation was not observed on bmMSCs. The expression of the cytokines investigated was not modulated significantly by PLL. We conclude that accelerated expansion of pMSCs in GMP-compliant media enriched by PLL reduces the expression of stemness marker CD146, but does not influence the expression of neuro- and myoregenerative cytokines.
Collapse
|
26
|
Craciunescu O, Seciu AM, Zarnescu O. In vitro and in vivo evaluation of a biomimetic scaffold embedding silver nanoparticles for improved treatment of oral lesions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:112015. [PMID: 33812634 DOI: 10.1016/j.msec.2021.112015] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 01/24/2023]
Abstract
BACKGROUND New materials are currently designed for efficient treatment of oral tissue lesions by guided tissue regeneration. The aim of this study was to develop a multifunctional 3D hybrid biomaterial consisting of extracellular matrix components, collagen, chondroitin 4-sulfate and fibronectin, functionalised with silver nanoparticles, intended to improve periodontitis treatment protocols. METHODS Structural observations were performed by autometallography, scanning and transmission electron microscopy. In vitro tests of 3D constructs of embedded gingival fibroblasts within hybrid biomaterial were performed by MTS and Live/Dead assays. Genotoxicity was assessed by comet assay. In vivo experiments using chick embryo chorioallantoic membrane (CAM) assay analysed the degradation and nanoparticles release, but also angiogenesis, new tissue formation in 3D constructs and the regenerative potential of the hybrid material. Biological activity was investigated in experimental models of inflamed THP-1 macrophages and oral specific bacterial cultures. RESULTS Light micrographs showed distribution of silver nanoparticles on collagen fibrils. Scanning electron micrographs revealed a microstructure with interconnected pores, which favoured cell adhesion and infiltration. Cell viability and proliferation were significantly higher within the 3D hybrid biomaterial than in 2D culture conditions, while absence of the hybrid material's genotoxic effect was found. In vivo experiments showed that the hybrid material was colonised by cells and blood vessels, initiating synthesis of new extracellular matrix. Besides the known effect of chondroitin sulfate, incorporated silver nanoparticles increased the anti-inflammatory activity of the hybrid biomaterial. The silver nanoparticles maintained their antibacterial activity even after embedding in the polymeric scaffold and inhibited the growth of F. nucleatum and P. gingivalis. CONCLUSION The novel biomimetic scaffold functionalised with silver nanoparticles presented regenerative, anti-inflammatory and antimicrobial potential for oral cavity lesions repair.
Collapse
Affiliation(s)
- Oana Craciunescu
- Department of Cellular and Molecular Biology, National Institute R&D for Biological Sciences, 296, Splaiul Independentei, 060031 Bucharest, Romania
| | - Ana-Maria Seciu
- Department of Cellular and Molecular Biology, National Institute R&D for Biological Sciences, 296, Splaiul Independentei, 060031 Bucharest, Romania; University of Bucharest, Faculty of Biology, Splaiul Independentei 91-95, 050095 Bucharest, Romania
| | - Otilia Zarnescu
- University of Bucharest, Faculty of Biology, Splaiul Independentei 91-95, 050095 Bucharest, Romania.
| |
Collapse
|
27
|
Deng F, Zhai W, Yin Y, Peng C, Ning C. Advanced protein adsorption properties of a novel silicate-based bioceramic: A proteomic analysis. Bioact Mater 2021; 6:208-218. [PMID: 32913929 PMCID: PMC7451930 DOI: 10.1016/j.bioactmat.2020.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/08/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
Silicate bioceramics have been shown to possess excellent cytocompatibility and osteogenic activity, but the exact mechanism is still unclear. Protein adsorption is the first event taking place at the biomaterial-tissue interface, which is vital to the subsequent cellular behavior and further influence the biomaterial-tissue interaction. In this work, the protein adsorption behavior of a novel CPS bioceramic was evaluated using the proteomics technology. The results showed that CPS adsorbed more amount and types of serum proteins than HA. FN1 and IGF1 proteins selected from proteomics results were validated by Western-blot experiment. Pathway analysis also revealed mechanistic insights how these absorbed proteins by CPS help mediate cell adhesion and promotes osteogenic activity. Firstly, the dramatically enhanced adsorption of FN1 could greatly promote cell adhesion and growth. Secondly, IGF1 was uniquely adsorbed on CPS bioceramic and IGF1 could activate Rap1 signaling pathway to promote cell adhesion. Thirdly, the increased adsorption of FN1, IGF1 and COL1A2 proteins on CPS explains its better ability on bone regeneration than HA. Fourthly, the increased adsorption of IGF1, CHAD, COL2A1 and THBS4 proteins on CPS explains its ability on cartilage formation. Lastly, the increased adsorption of immunological related proteins on CPS may also play a positive role in bone regeneration. In addition, CPS had a much better cell adhesion ability than HA, proving that more adsorbed proteins really had a positive effect on cell behavior. The more adsorbed proteins on CPS than HA might indicated a better bone regeneration rate at early stage of implantation.
Collapse
Affiliation(s)
- Fanyan Deng
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Wanyin Zhai
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Yue Yin
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, 201210, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, 201210, China
| | - Congqin Ning
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, 200234, China
| |
Collapse
|
28
|
Liu C, Sun J. Impact of Marine-Based Biomaterials on the Immunoregulatory Properties of Bone Marrow-Derived Mesenchymal Stem Cells: Potential Use of Fish Collagen in Bone Tissue Engineering. ACS OMEGA 2020; 5:28360-28368. [PMID: 33163819 PMCID: PMC7643310 DOI: 10.1021/acsomega.0c04360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
A key issue in the field of tissue engineering and stem cell therapy is immunological rejection after the implantation of allogeneic bone marrow-derived mesenchymal stem cells (BMSCs). In addition, maintaining the immunoregulatory function of BMSCs is critical to achieving tissue repair. In recent years, scientists have become interested in fish collagen because of its unique osteoinductive activity. However, it is still unclear whether osteogenically differentiated BMSCs induced by fish collagen maintain their immunoregulatory functions. To address this question, BMSCs were isolated from 8-week-old male BALB/c mice, and a noncontact coculture model was established consisting of macrophages and BMSCs treated with hydrolyzed fish collagen (HFC). Cell proliferation of the macrophages was determined by MTT. The gene and protein expression levels of the M1 and M2 macrophage markers were measured by real-time PCR and enzyme-linked immunosorbent assay (ELISA). To study the role of TNF-α-induced gene/protein 6 (TSG-6), TSG-6 was targeted by short interfering RNA (siRNA) in BMSCs, then the osteogenic differentiation ability of the BMSCs was examined by western blotting. The mRNA expression levels of interleukin-10 (IL-10), CCL22 (a macrophage-derived chemokine), tumor necrosis factor α (TNF-α), and interleukin-12 (IL-12), and the protein expression levels of arginase-1 (Arg-1) and inducible nitric oxide synthase (iNOS) of macrophages cocultured with TSG-6-siRNA-BMSCs+HFC were detected by real-time PCR and western blotting, respectively. The results showed that the osteogenically differentiated BMSCs induced by HFC did not affect the proliferation of macrophages. Osteogenically differentiated BMSCs induced by HFC promoted the expression of M2 macrophage markers IL-10 and CCL22, while HFC inhibited the expression of M1 macrophage markers, including TNF-α and IL-12. The TSG-6 knockdown led to a decrease in the production of TSG-6 without impairing the expression of bone sialoprotein (BSP), osteocalcin (OCN), and Runt-related transcription factor 2 (RUNX2) by BMSCs. TSG-6 silencing significantly counteracted the effect of HFC, and the expression of IL-10, CCL22, and Arg-1 were all decreased in the macrophages cocultured with TSG-6-siRNA-BMSCs+HFC, while that of TNF-α, IL-12, and iNOS were increased relative to the BMSCs+HFC group. The data demonstrated that osteogenically differentiated BMSCs induced by fish collagen retained their immunomodulatory functions. This study provides an additional scientific basis for future applications of fish collagen as an osteogenic component in the fields of tissue engineering and stem cell therapy.
Collapse
|
29
|
Fibrin as a Multipurpose Physiological Platform for Bone Tissue Engineering and Targeted Delivery of Bioactive Compounds. Pharmaceutics 2019; 11:pharmaceutics11110556. [PMID: 31661853 PMCID: PMC6920828 DOI: 10.3390/pharmaceutics11110556] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/18/2019] [Accepted: 10/18/2019] [Indexed: 12/14/2022] Open
Abstract
Although bone graft is still considered as the gold standard method, bone tissue engineering offers promising alternatives designed to mimic the extracellular matrix (ECM) and to guide bone regeneration process. In this attempt, due to their similarity to the ECM and their low toxicity/immunogenicity properties, growing attention is paid to natural polymers. In particular, considering the early critical role of fracture hematoma for bone healing, fibrin, which constitutes blood clot, is a candidate of choice. Indeed, in addition to its physiological roles in bone healing cascade, fibrin biochemical characteristics make it suitable to be used as a multipurpose platform for bioactive agents’ delivery. Thus, taking advantage of these key assets, researchers and clinicians have the opportunity to develop composite systems that might further improve bone tissue reconstruction, and more generally prevent/treat skeletal disorders.
Collapse
|
30
|
Kado T, Aita H, Ichioka Y, Endo K, Furuichi Y. Chemical modification of pure titanium surfaces to enhance the cytocompatibility and differentiation of human mesenchymal stem cells. Dent Mater J 2019; 38:1026-1035. [PMID: 31582594 DOI: 10.4012/dmj.2018-257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The aim of this study was to improve the cytocompatibility and differentiation of human bone marrow-derived mesenchymal stem cells on the surface of titanium implants by immobilizing biofunctional molecules on their surface. Gly-Arg-Gly-Asp-Ser (GRGDS) peptides, human plasma fibronectin (pFN), or type I collagen from calf skin (Col) was covalently immobilized on the titanium surfaces. Twice as many cells attached to the Col- and pFN-immobilized titanium surfaces than attached to the as-polished surface control. The ALP activity of the cells, as well as the mineralized nodule formation, was significantly higher on the Col- and pFN-immobilized titanium surfaces than on the as-polished surfaces. These results indicate that the immobilization of biofunctional molecules such as Col and pFN on titanium surfaces enhances the attachment, spreading, proliferation, and differentiation of human bone marrow-derived mesenchymal stem cells, which may lead to a more rapid bone-titanium integration.
Collapse
Affiliation(s)
- Takashi Kado
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido
| | - Hideki Aita
- Division of Geriatric Dentistry, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido
| | - Yuki Ichioka
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido
| | - Kazuhiko Endo
- Division of Biomaterials and Bioengineering, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido
| | - Yasushi Furuichi
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido
| |
Collapse
|
31
|
Halib N, Ahmad I, Grassi M, Grassi G. The remarkable three-dimensional network structure of bacterial cellulose for tissue engineering applications. Int J Pharm 2019; 566:631-640. [PMID: 31195074 DOI: 10.1016/j.ijpharm.2019.06.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/21/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022]
Abstract
Cellulose is a natural homopolymer, composed of β-1,4- anhydro-d-glucopyranose units. Unlike plant cellulose, bacterial cellulose (BC), obtained from species belonging to the genera of Acetobacter, Rhizobium, Agrobacterium, and Sarcina through various cultivation methods and techniques, is produced in its pure form. BC is produced in the form of gel-like, never dry sheet with tremendous mechanical properties. Containing up to 99% of water, BC hydrogel is considered biocompatible thus finding robust applications in the health industry. Moreover, BC three-dimensional structure closely resembles the extracellular matrix (ECM) of living tissue. In this review, we focus on the porous BC morphology particularly suited to host oxygen and nutrients thus providing conducive environment for cell growth and proliferation. The remarkable BC porous morphology makes this biological material a promising templet for the generation of 3D tissue culture and possibly for tissue-engineered scaffolds.
Collapse
Affiliation(s)
- Nadia Halib
- Department of Basic Sciences & Oral Biology, Faculty of Dentistry, Universiti Sains Islam Malaysia, Kuala Lumpur 55100, Malaysia.
| | - Ishak Ahmad
- Centre for Advanced Materials and Renewable Resources, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor Darul Ehsan, Malaysia
| | - Mario Grassi
- Department of Engineering and Architecture, Trieste University, via Valerio 6, I-34127 Trieste, Italy
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy
| |
Collapse
|
32
|
Sun R, Xu S, Wang Z. Rat sinus mucosa- and periosteum-derived exosomes accelerate osteogenesis. J Cell Physiol 2019; 234:21947-21961. [PMID: 31074002 DOI: 10.1002/jcp.28758] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/28/2019] [Accepted: 04/11/2019] [Indexed: 12/30/2022]
Abstract
Guided bone regeneration (GBR) is commonly used for alveolar bone augmentation. The paracrine mechanism in the field of bone tissue engineering has been emphasized in recent years and exosomes are considered to have the potential of promoting osteogenesis. We aimed to study the influence of sinus mucosa and periosteum on bone regeneration through paracrine stimulation, especially via exosomes, and compare the differences between them. Here, we report that conditioned medium (CM) from sinus mucosa-derived cells (SMCs) and periosteum-derived cells (PCs) and the isolated exosomes enhanced the proliferation, migration and osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BM-MSCs) in vitro. A rat model of femoral bone defects was used to demonstrate that the exosomes derived from SMCs (SMC-Exos) and PCs (PC-Exos) can accelerate bone formation in vivo. Furthermore, we present a preliminary discussion of the possible functional components involved in the effects of SMC-Exos and PC-Exos on bone regeneration. In conclusion, these results demonstrated that the sinus mucosa and periosteum can accelerate osteogenesis through paracrine effects and the exosomes play important roles in this process.
Collapse
Affiliation(s)
- Ruinan Sun
- Department of Oral Implant, School of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Hospital of Stomatology, Tongji University, Shanghai, China
| | - Shuyu Xu
- Department of Oral Implant, School of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Hospital of Stomatology, Tongji University, Shanghai, China
| | - Zuolin Wang
- Department of Oral Implant, School of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Hospital of Stomatology, Tongji University, Shanghai, China
| |
Collapse
|
33
|
Lerman MJ, Muramoto S, Arumugasaamy N, Van Order M, Lembong J, Gerald AG, Gillen G, Fisher JP. Development of surface functionalization strategies for 3D-printed polystyrene constructs. J Biomed Mater Res B Appl Biomater 2019; 107:2566-2578. [PMID: 30821930 DOI: 10.1002/jbm.b.34347] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/18/2019] [Accepted: 02/10/2019] [Indexed: 01/17/2023]
Abstract
There is a growing interest in 3D printing to fabricate culture substrates; however, the surface properties of the scaffold remain pertinent to elicit targeted and expected cell responses. Traditional 2D polystyrene (PS) culture systems typically require surface functionalization (oxidation) to facilitate and encourage cell adhesion. Determining the surface properties which enhance protein adhesion from media and cellular extracellular matrix (ECM) production remains the first step to translating 2D PS systems to a 3D culture surface. Here we show that the presence of carbonyl groups to PS surfaces correlated well with successful adhesion of ECM proteins and sustaining ECM production of deposited human mesenchymal stem cells, if the surface has a water contact angle between 50° and 55°. Translation of these findings to custom-fabricated 3D PS scaffolds reveals carbonyl groups continued to enhance spreading and growth in 3D culture. Cumulatively, these data present a method for 3D printing PS and the design considerations required for understanding cell-material interactions. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B:2566-2578, 2019.
Collapse
Affiliation(s)
- Max J Lerman
- Department of Materials Science and Engineering, University of Maryland, College Park, Maryland.,Surface and Trace Chemical Analysis Group, Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland.,Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
| | - Shin Muramoto
- Surface and Trace Chemical Analysis Group, Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - Navein Arumugasaamy
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland.,Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia
| | - Michael Van Order
- Department of Materials Science and Engineering, University of Maryland, College Park, Maryland
| | - Josephine Lembong
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland
| | - Anushka G Gerald
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland
| | - Greg Gillen
- Surface and Trace Chemical Analysis Group, Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - John P Fisher
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland
| |
Collapse
|
34
|
Stewart CAC, Akhavan B, Hung J, Bao S, Jang JH, Wise SG, Bilek MMM. Multifunctional Protein-Immobilized Plasma Polymer Films for Orthopedic Applications. ACS Biomater Sci Eng 2018; 4:4084-4094. [DOI: 10.1021/acsbiomaterials.8b00954] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Callum A. C. Stewart
- School of Physics, University of Sydney, Physics Road, Camperdown, NSW 2006, Australia
- Heart Research Institute, 7 Eliza Street, Newtown, New South Wales 2042, Australia
- Charles Perkins Centre, University of Sydney, Camperdown NSW 2006, Australia
| | - Behnam Akhavan
- School of Physics, University of Sydney, Physics Road, Camperdown, NSW 2006, Australia
- Heart Research Institute, 7 Eliza Street, Newtown, New South Wales 2042, Australia
- School of Aerospace Mechanical and Mechatronic Engineering, University of Sydney, Camperdown, NSW 2006, Australia
| | - Juichien Hung
- Heart Research Institute, 7 Eliza Street, Newtown, New South Wales 2042, Australia
| | - Shisan Bao
- Charles Perkins Centre, University of Sydney, Camperdown NSW 2006, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW 2006, Australia
| | - Jun-Hyeog Jang
- School of Medicine, Inha University, Incheon 400−712, Korea
| | - Steven G. Wise
- Heart Research Institute, 7 Eliza Street, Newtown, New South Wales 2042, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW 2006, Australia
| | - Marcela M. M. Bilek
- School of Physics, University of Sydney, Physics Road, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, University of Sydney, Camperdown NSW 2006, Australia
- School of Aerospace Mechanical and Mechatronic Engineering, University of Sydney, Camperdown, NSW 2006, Australia
- Sydney Nano Institute, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
35
|
Nie JM, Li HF. Therapeutic effects of Salvia miltiorrhiza injection combined with telmisartan in patients with diabetic nephropathy by influencing collagen IV and fibronectin: A case-control study. Exp Ther Med 2018; 16:3405-3412. [PMID: 30233688 PMCID: PMC6143830 DOI: 10.3892/etm.2018.6654] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 08/01/2018] [Indexed: 01/11/2023] Open
Abstract
Involvement of collagen IV (ColIV) and fibronectin (FN) in the occurrence and development of diabetic nephropathy (DN) and the effects of telmisartan and Salvia miltiorrhiza injection in the treatment of the patients were investigated. Two hundred and fifty-eight patients with stage IV DN were selected as the case group, and another 110 normal healthy subjects were incorporated as the control group. Involved patients were subdivided into different groups according to different treatment therapies; patients in the telmisartan group (T group) were given oral telmisartan; patients in the Salvia miltiorrhiza injection + telmisartan (S + T group) were administered with Salvia miltiorrhiza injection combined with telmisartan treatment, and there was a group of patients who received no intervention as the placebo group. After intervention, levels of glycemic indexes and renal damage indexes indicated downwards trends both in the T group and the S + T group when compared to the placebo group; besides, levels in the S + T group were much lower than those in the T group (all P<0.05). Additionally, in comparison among the above three intervention groups, differences in the fasting blood glucose, 2 h post-prandial blood glucose, glycosylated hemoglobin, blood urea nitrogen, serum creatinine and urinary albumin excretion rate were significant after treatment (all P<0.05). Further, before intervention, both Co1IV and FN in the urine were increased in the case group compared to the control group (all P<0.05). After intervention, both levels were apparently decreased. There were remarkable differences of Co1IV and FN levels in the urine when compared among three different intervention groups after treatment (P<0.05). Increased ColIV and FN levels may be partially responsible for the development of DN. Salvia miltiorrhiza injection with telmisartan have beneficial synergistic effects for DN patients through attenuating the increase in ColIV and FN, reversing hyperglycemia state and postponing ultrastructure changes of glomerular basement membrane.
Collapse
Affiliation(s)
- Jie-Ming Nie
- Department of Internal Medicine, Τhe Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Hai-Feng Li
- Department of Pharmaceutical Analysis, ALK-Abello A/S Guangzhou Office, Guangzhou, Guangdong 510620, P.R. China
| |
Collapse
|
36
|
Yu NH, Chun SY, Ha YS, Kim HT, Kim DH, Kim J, Chung JW, Lee JN, Song PH, Yoo ES, Kim BS, Kwon TG. Optimal Stem Cell Transporting Conditions to Maintain Cell Viability and Characteristics. Tissue Eng Regen Med 2018; 15:639-647. [PMID: 30603585 DOI: 10.1007/s13770-018-0133-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The preservation of stem cell viability and characteristics during cell transport from the bench to patients can significantly affect the success of cell therapy. Factors such as suspending medium, time, temperature, cell density, and container type could be considered for transport conditions. METHODS To establish optimal conditions, human amniotic fluid stem cells' (AFSCs) viabilities were analyzed under different media {DMEM(H), DMEM/F-12, K-SFM, RPMI 1640, α-MEM, DMEM(L), PBS or saline}, temperature (4, 22 or 37 °C), cell density (1 × 107 cells were suspended in 0.1, 0.5, 1.0 or 2.0 mL of medium) and container type (plastic syringe or glass bottle). After establishing the transport conditions, stem cell characteristics of AFSCs were compared to freshly prepared cells. RESULTS Cells transported in DMEM(H) showed relatively higher viability than other media. The optimized transport temperature was 4 °C, and available transport time was within 12 h. A lower cell density was associated with a better survival rate, and a syringe was selected as a transport container because of its clinical convenience. In compare of stem cell characteristics, the transported cells with established conditions showed similar potency as the freshly prepared cells. CONCLUSION Our findings can provide a foundation to optimization of conditions for stem cell transport.
Collapse
Affiliation(s)
- Na-Hee Yu
- 1Biomedical Research Institute, Kyungpook National University Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea
| | - So Young Chun
- 1Biomedical Research Institute, Kyungpook National University Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea
| | - Yun-Sok Ha
- 2Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea.,3Department of Urology, Kyungpook National University Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea
| | - Hyun Tae Kim
- 2Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea.,4Department of Urology, Kyungpook National University Chilgok Hospital, 807 Hogukro, Buk-gu, Daegu, 41404 Korea
| | - Dae Hwan Kim
- 5Department of Laboratory Animal Research Support Team, Yeungnam University Medical Center, 170 Hyunchung-ro, Nam-gu, Daegu, 42415 Korea
| | - Jeongshik Kim
- Department of Pathology, Central Hospital, 480 Munsu-ro, Nam-gu, Ulsan, 44667 Korea
| | - Jae-Wook Chung
- 2Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea.,4Department of Urology, Kyungpook National University Chilgok Hospital, 807 Hogukro, Buk-gu, Daegu, 41404 Korea
| | - Jun Nyung Lee
- 2Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea.,4Department of Urology, Kyungpook National University Chilgok Hospital, 807 Hogukro, Buk-gu, Daegu, 41404 Korea
| | - Phil Hyun Song
- 7Department of Urology, College of Medicine, Yeungnam University, 170 Hyunchung-ro, Nam-gu, Daegu, 42415 Korea
| | - Eun Sang Yoo
- 2Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea.,3Department of Urology, Kyungpook National University Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea
| | - Bum Soo Kim
- 2Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea.,3Department of Urology, Kyungpook National University Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea
| | - Tae Gyun Kwon
- 2Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea.,4Department of Urology, Kyungpook National University Chilgok Hospital, 807 Hogukro, Buk-gu, Daegu, 41404 Korea
| |
Collapse
|
37
|
Montalbano G, Toumpaniari S, Popov A, Duan P, Chen J, Dalgarno K, Scott WE, Ferreira AM. Synthesis of bioinspired collagen/alginate/fibrin based hydrogels for soft tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 91:236-246. [PMID: 30033251 DOI: 10.1016/j.msec.2018.04.101] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 03/13/2018] [Accepted: 04/28/2018] [Indexed: 12/22/2022]
Abstract
Hydrogels based on natural polymers offer a range of properties to mimic the native extracellular matrix, and provide microenvironments to preserve cellular function and encourage tissue formation. A tri-component hydrogel using collagen, alginate and fibrin (CAF) was developed and investigated at three collagen concentrations for application as a functional extracellular matrix analogue. Physical-chemical characterization of CAF hydrogels demonstrated a thermo-responsive crosslinking capacity at physiological conditions with stiffness similar to native soft tissues. CAF hydrogels were also assessed for cytocompatibility using L929 murine fibroblasts, pancreatic MIN6 β-cells and human mesenchymal stem cells (hMSCs); and demonstrated good cell viability, proliferation and metabolic activity after 7 days of in vitro culture. CAF hydrogels, especially with 2.5% w/v collagen, increased alkaline phosphatase production in hMSCs indicating potential for the promotion of osteogenic activity. Moreover, CAF hydrogels also increased metabolic activity of MIN6 β-cells and promoted the reconstitution of spherical pseudoislets with sizes ranging between 50 and 150 μm at day 7, demonstrating potential in diabetic therapeutic applications.
Collapse
Affiliation(s)
- G Montalbano
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; Department of Applied Science and Technology, Politecnico di Torino, Turin 10129, Italy
| | - S Toumpaniari
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; Cambridge Centre for Medical Materials, University of Cambridge, Cambridge CB3 0FS, UK
| | - A Popov
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; UCL Cancer Institute, University College London, London WC1E 6BT, UK
| | - P Duan
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
| | - J Chen
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
| | - K Dalgarno
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
| | - W E Scott
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK
| | - A M Ferreira
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK.
| |
Collapse
|
38
|
Developmental Pathways Pervade Stem Cell Responses to Evolving Extracellular Matrices of 3D Bioprinted Microenvironments. Stem Cells Int 2018; 2018:4809673. [PMID: 29765414 PMCID: PMC5896227 DOI: 10.1155/2018/4809673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/10/2017] [Accepted: 12/10/2017] [Indexed: 12/17/2022] Open
Abstract
Developmental studies and 3D in vitro model systems show that the production and engagement of extracellular matrix (ECM) often precede stem cell differentiation. Yet, unclear is how the ECM triggers signaling events in sequence to accommodate multistep process characteristic of differentiation. Here, we employ transcriptome profiling and advanced imaging to delineate the specificity of ECM engagement to particular differentiation pathways and to determine whether specificity in this context is a function of long-term ECM remodeling. To this end, human mesenchymal stem cells (hMSCs) were cultured in 3D bioprinted prisms created from ECM proteins and associated controls. We found that exogenous ECM provided in 3D microenvironments at early time points impacts on the composition of microenvironments at later time points and that each evolving 3D microenvironment is uniquely poised to promote stem cell differentiation. Moreover, 2D cultures undergo minimal ECM remodeling and are ill-equipped to stimulate pathways associated with development.
Collapse
|
39
|
Störmann P, Kupsch J, Kontradowitz K, Leiblein M, Verboket R, Seebach C, Marzi I, Henrich D, Nau C. Cultivation of EPC and co-cultivation with MSC on β-TCP granules in vitro is feasible without fibronectin coating but influenced by scaffolds' design. Eur J Trauma Emerg Surg 2018. [PMID: 29523894 DOI: 10.1007/s00068-018-0935-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Meanwhile, the osteoconductive properties of frequently used synthetic bone grafts can be improved by the use of osteoinductive cells and growth factors. Nevertheless, the cultivation of endothelial progenitor cells (EPC) seems to be difficult and requires a pre-conditioning of the scaffolds with fibronectin. Additionally, the influence of the scaffolds' design on cell cultivation is not fully elucidated. METHODS As scaffold, a commercially available β-tricalcium phosphate was used. 5 × 105 EPC, or 5 × 105 MSC or a combination of each 2.5 × 105 cells was seeded onto the granules. We investigated seeding efficiency, cell morphology, cell metabolism, adherence, apoptosis and gene expression of EPC and MSC in this in vitro study on days 2, 6 and 10. RESULTS Total number of adherent cells was higher on the β-TCP without fibronectin coating. The number of cells in all approaches significantly declined when a solid β-TCP was used. Metabolic activity of MSC was comparable throughout the scaffolds and increased until day 10. Additionally, the amount of supernatants VEGF was higher for MSC than for EPC. DISCUSSION Our results demonstrate that a coating of the scaffold for successful cultivation of EPC in vitro is not necessary. Furthermore, our study showed that structural differences of the scaffolds significantly influenced cell adherence and metabolic activity. Thereby, the influence on EPC seems to be higher than on MSC.
Collapse
Affiliation(s)
- Philipp Störmann
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Johann Wolfgang Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
| | - Juliane Kupsch
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Johann Wolfgang Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Kerstin Kontradowitz
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Johann Wolfgang Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Maximilian Leiblein
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Johann Wolfgang Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - René Verboket
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Johann Wolfgang Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Caroline Seebach
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Johann Wolfgang Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Johann Wolfgang Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Dirk Henrich
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Johann Wolfgang Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Christoph Nau
- Department of Trauma, Hand and Reconstructive Surgery, Hospital of the Johann Wolfgang Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| |
Collapse
|
40
|
Zhou L, Chen X, Liu T, Zhu C, Si M, Jargstorf J, Li M, Pan G, Gong Y, Luo ZP, Yang H, Pei M, He F. SIRT1-dependent anti-senescence effects of cell-deposited matrix on human umbilical cord mesenchymal stem cells. J Tissue Eng Regen Med 2018; 12:e1008-e1021. [PMID: 28107614 PMCID: PMC9805355 DOI: 10.1002/term.2422] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/07/2016] [Accepted: 01/17/2017] [Indexed: 01/03/2023]
Abstract
Human umbilical cord-derived mesenchymal stem cells (UC-MSCs) are considered an attractive cell source for tissue regeneration. However, environmental oxidative stress can trigger premature senescence in MSCs and thus compromises their regenerative potential. Extracellular matrix (ECM) derived from MSCs has been shown to facilitate cell proliferation and multi-lineage differentiation. This investigation evaluated the effect of cell-deposited decellularized ECM (DECM) on oxidative stress-induced premature senescence in UC-MSCs. Sublethal dosages of H2 O2 , ranging from 50 μm to 200 μm, were used to induce senescence in MSCs. We found that DECM protected UC-MSCs from oxidative stress-induced premature senescence. When treated with H2 O2 at the same concentration, cell proliferation of DECM-cultured UC-MSCs was twofold higher than those on standard tissue culture polystyrene (TCPS). After exposure to 100 μm H2 O2 , fewer senescence-associated β-galactosidase-positive cells were observed on DECM than those on TCPS (17.6 ± 4.0% vs. 60.4 ± 6.2%). UC-MSCs cultured on DECM also showed significantly lower levels of senescence-related regulators, such as p16INK4α and p21. Most importantly, DECM preserved the osteogenic differentiation potential of UC-MSCs with premature senescence. The underlying molecular mechanisms involved the silent information regulator type 1 (SIRT1)-dependent signalling pathway, confirmed by the fact that the SIRT1 inhibitor nicotinamide counteracted the DECM-mediated anti-senescent effect. Collagen type I, rather than fibronectin, partially contributed to the protective effect of decellularized matrix. These findings provide a new strategy of using stem cell-deposited matrix to overcome the challenge of cellular senescence and to facilitate the clinical application of MSCs in regenerative medicine. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Long Zhou
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xi Chen
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, China
| | - Tao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Caihong Zhu
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Michelle Si
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Departments of Biology and Chemistry, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Joseph Jargstorf
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Biology, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Mao Li
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guoqing Pan
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Yihong Gong
- School of Engineering, Sun Yat-sen University, Guangzhou, China
| | - Zong-Ping Luo
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huilin Yang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, Morgantown, WV, USA
| | - Fan He
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
41
|
Goetzke R, Franzen J, Ostrowska A, Vogt M, Blaeser A, Klein G, Rath B, Fischer H, Zenke M, Wagner W. Does soft really matter? Differentiation of induced pluripotent stem cells into mesenchymal stromal cells is not influenced by soft hydrogels. Biomaterials 2017; 156:147-158. [PMID: 29197223 DOI: 10.1016/j.biomaterials.2017.11.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/21/2017] [Indexed: 01/22/2023]
Abstract
Induced pluripotent stem cells (iPSCs) can be differentiated toward mesenchymal stromal cells (MSCs), but this transition remains incomplete. It has been suggested that matrix elasticity directs cell-fate decisions. Therefore, we followed the hypothesis that differentiation of primary MSCs and generation of iPSC-derived MSCs (iMSCs) is supported by a soft matrix of human platelet lysate (hPL-gel). We demonstrate that this fibrin-based hydrogel supports growth of primary MSCs with pronounced deposition of extracellular matrix, albeit it hardly impacts on gene expression profiles or in vitro differentiation of MSCs. Furthermore, iPSCs can be effectively differentiated toward MSC-like cells on the hydrogel. Unexpectedly, this complex differentiation process is not affected by the substrate: iMSCs generated on tissue culture plastic (TCP) or hPL-gel have the same morphology, immunophenotype, differentiation potential, and gene expression profiles. Moreover, global DNA methylation patterns are essentially identical in iMSCs generated on TCP or hPL-gel, indicating that they are epigenetically alike. Taken together, hPL-gel provides a powerful matrix that supports growth and differentiation of primary MSCs and iMSCs - but this soft hydrogel does not impact on lineage-specific differentiation.
Collapse
Affiliation(s)
- Roman Goetzke
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Julia Franzen
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Alina Ostrowska
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Michael Vogt
- Interdisciplinary Center for Clinical Research IZKF Aachen, RWTH Aachen, University Medical School, Aachen, Germany
| | - Andreas Blaeser
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Gerd Klein
- Center for Medical Research, Department of Medicine II, University of Tübingen, Tübingen, Germany
| | - Björn Rath
- Department of Orthopedics, RWTH Aachen University Medical School, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Martin Zenke
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany; Institute for Biomedical Engineering - Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Wolfgang Wagner
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany; Institute for Biomedical Engineering - Cell Biology, RWTH Aachen University Medical School, Aachen, Germany.
| |
Collapse
|
42
|
Enhanced Cell Growth of Adipocyte-Derived Mesenchymal Stem Cells Using Chemically-Defined Serum-Free Media. Int J Mol Sci 2017; 18:ijms18081779. [PMID: 28813021 PMCID: PMC5578168 DOI: 10.3390/ijms18081779] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 08/12/2017] [Accepted: 08/14/2017] [Indexed: 12/17/2022] Open
Abstract
The multipotency and anti-inflammatory effects of mesenchymal stem cells (MSCs) make them attractive for cell therapy in regenerative medicine. A large number of MSCs is required for efficient therapy owing to the low homing efficiency of MSCs to target sites. Furthermore, owing to limitations in obtaining sufficient amounts of MSCs, in vitro expansion of MSCs that preserves their differentiation and proliferative potential is essential. The animal factor included in culture media also limits clinical application. In this study, adipose-derived MSCs showed a significantly higher proliferation rate in STK2, a chemically-defined medium, than in DMEM/FBS. The expression of MSC surface markers was increased in the culture using STK2 compared to that using DMEM/FBS. Tri-lineage differentiation analyses showed that MSCs cultured in STK2 were superior to those cultured in DMEM/FBS. In addition, MSCs cultured in STK2 showed a reduced senescence rate, small and homogenous cell size, and were more genetically stable compared to those cultured in DMEM/FBS. Furthermore, secretome analysis showed that the expression of factors related to proliferation/migration, anti-inflammation, and differentiation were increased in STK2 culture medium compared to DMEM/FBS. Taken together, these results suggest that culture using STK2 medium offers many advantages through which it is possible to obtain safer, superior, and larger numbers of MSCs.
Collapse
|
43
|
Noori A, Ashrafi SJ, Vaez-Ghaemi R, Hatamian-Zaremi A, Webster TJ. A review of fibrin and fibrin composites for bone tissue engineering. Int J Nanomedicine 2017; 12:4937-4961. [PMID: 28761338 PMCID: PMC5516781 DOI: 10.2147/ijn.s124671] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Tissue engineering has emerged as a new treatment approach for bone repair and regeneration seeking to address limitations associated with current therapies, such as autologous bone grafting. While many bone tissue engineering approaches have traditionally focused on synthetic materials (such as polymers or hydrogels), there has been a lot of excitement surrounding the use of natural materials due to their biologically inspired properties. Fibrin is a natural scaffold formed following tissue injury that initiates hemostasis and provides the initial matrix useful for cell adhesion, migration, proliferation, and differentiation. Fibrin has captured the interest of bone tissue engineers due to its excellent biocompatibility, controllable biodegradability, and ability to deliver cells and biomolecules. Fibrin is particularly appealing because its precursors, fibrinogen, and thrombin, which can be derived from the patient's own blood, enable the fabrication of completely autologous scaffolds. In this article, we highlight the unique properties of fibrin as a scaffolding material to treat bone defects. Moreover, we emphasize its role in bone tissue engineering nanocomposites where approaches further emulate the natural nanostructured features of bone when using fibrin and other nanomaterials. We also review the preparation methods of fibrin glue and then discuss a wide range of fibrin applications in bone tissue engineering. These include the delivery of cells and/or biomolecules to a defect site, distributing cells, and/or growth factors throughout other pre-formed scaffolds and enhancing the physical as well as biological properties of other biomaterials. Thoughts on the future direction of fibrin research for bone tissue engineering are also presented. In the future, the development of fibrin precursors as recombinant proteins will solve problems associated with using multiple or single-donor fibrin glue, and the combination of nanomaterials that allow for the incorporation of biomolecules with fibrin will significantly improve the efficacy of fibrin for numerous bone tissue engineering applications.
Collapse
Affiliation(s)
- Alireza Noori
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran
| | | | - Roza Vaez-Ghaemi
- Department of Chemical and Biological Engineering, Faculty of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | | | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
44
|
Akbarzadeh M, Rahbarghazi R, Nabat E, Movassaghpour AA, Shanehbandi D, Faramarzian Azimi Maragheh B, Matluobi D, Barazvan B, Kazemi M, Samadi N, Nouri M. The impact of different extracellular matrices on melatonin effect in proliferation and stemness properties of ovarian cancer cells. Biomed Pharmacother 2017; 87:288-295. [DOI: 10.1016/j.biopha.2016.12.119] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/25/2016] [Accepted: 12/27/2016] [Indexed: 12/24/2022] Open
|
45
|
Jafari A, Qanie D, Andersen TL, Zhang Y, Chen L, Postert B, Parsons S, Ditzel N, Khosla S, Johansen HT, Kjærsgaard-Andersen P, Delaisse JM, Abdallah BM, Hesselson D, Solberg R, Kassem M. Legumain Regulates Differentiation Fate of Human Bone Marrow Stromal Cells and Is Altered in Postmenopausal Osteoporosis. Stem Cell Reports 2017; 8:373-386. [PMID: 28162997 PMCID: PMC5312427 DOI: 10.1016/j.stemcr.2017.01.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/03/2017] [Accepted: 01/04/2017] [Indexed: 12/21/2022] Open
Abstract
Secreted factors are a key component of stem cell niche and their dysregulation compromises stem cell function. Legumain is a secreted cysteine protease involved in diverse biological processes. Here, we demonstrate that legumain regulates lineage commitment of human bone marrow stromal cells and that its expression level and cellular localization are altered in postmenopausal osteoporotic patients. As shown by genetic and pharmacological manipulation, legumain inhibited osteoblast (OB) differentiation and in vivo bone formation through degradation of the bone matrix protein fibronectin. In addition, genetic ablation or pharmacological inhibition of legumain activity led to precocious OB differentiation and increased vertebral mineralization in zebrafish. Finally, we show that localized increased expression of legumain in bone marrow adipocytes was inversely correlated with adjacent trabecular bone mass in a cohort of patients with postmenopausal osteoporosis. Our data suggest that altered proteolytic activity of legumain in the bone microenvironment contributes to decreased bone mass in postmenopausal osteoporosis. Legumain determines differentiation fate of BMSCs in vitro and in vivo Legumain regulates BMSC proliferation independent of its enzymatic activity Inhibition of legumain leads to precocious bone formation in zebrafish Legumain is overexpressed in bone marrow adipocytes of osteoporotic patients
Collapse
Affiliation(s)
- Abbas Jafari
- Department of Cellular and Molecular Medicine, Danish Stem Cell Center (DanStem), University of Copenhagen, 2200 Copenhagen, Denmark; Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark
| | - Diyako Qanie
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark
| | - Thomas L Andersen
- Department of Clinical Cell Biology, Vejle/ Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, 7100, Vejle, Denmark
| | - Yuxi Zhang
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Li Chen
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark
| | - Benno Postert
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Stuart Parsons
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Nicholas Ditzel
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark
| | - Sundeep Khosla
- Endocrine Research Unit, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | - Jean-Marie Delaisse
- Department of Clinical Cell Biology, Vejle/ Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, 7100, Vejle, Denmark
| | - Basem M Abdallah
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark; Department of Biological Sciences, College of Science, King Faisal University, Hofuf 6996, Saudi Arabia
| | - Daniel Hesselson
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St Vincent's Clinical School, UNSW Australia, Sydney, NSW 2010, Australia
| | - Rigmor Solberg
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, 0363 Oslo, Norway
| | - Moustapha Kassem
- Department of Cellular and Molecular Medicine, Danish Stem Cell Center (DanStem), University of Copenhagen, 2200 Copenhagen, Denmark; Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark; Stem Cell Unit, Department of Anatomy, Faculty of Medicine, King Saud University, Riyadh 12372, Saudi Arabia.
| |
Collapse
|
46
|
Adsorbed Fibrinogen stimulates TLR-4 on monocytes and induces BMP-2 expression. Acta Biomater 2017; 49:296-305. [PMID: 27856281 DOI: 10.1016/j.actbio.2016.11.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/26/2016] [Accepted: 11/14/2016] [Indexed: 11/22/2022]
Abstract
Modulation of inflammatory responses to implanted biomaterials towards tissue regeneration has gained prominence as an innovative tissue engineering strategy. Recent in vitro and in vivo studies showed that Fibrinogen (Fg) adsorbed to Chitosan (Ch) substrates modulates immune cell responses, enhances the production of osteogenic factors by monocytes/macrophages and promotes bone regeneration, but the mechanisms involved remain poorly understood. Thus, the present work was conducted to clarify the molecular mechanisms of interaction between primary human monocytes and the above substrates. Cell surface expression of TLR-4 was significantly downregulated in the presence of pre-adsorbed Fg, when compared to Ch control, indicating an interaction via this receptor. The same substrate triggered MAPK activation, specifically the ERK 1/2 and JNK pathways. Importantly, both ERK 1/2 and JNK phosphorylation were reduced when TLR-4 signalling was blocked using a specific pharmacological inhibitor. Functionally, adsorbed Fg induced production of the potent osteogenic mediator BMP-2 by monocytes, while TLR-4 inhibition resulted in a significant decrease of BMP-2 mRNA and protein levels, in response to Fg stimulation. Overall, our data reveals that adsorbed Fg exerts a pro-osteogenic effect on human monocytes through its interaction with TLR-4 and subsequent production of BMP-2, elucidating two key aspects of the immunomodulatory action of adsorbed Fg in bone regeneration. STATEMENT OF SIGNIFICANCE Recent studies showed that when Fibrinogen (Fg) is used to modify Chitosan (Ch) substrates, it modulates the immune response, enhances production of osteogenic factors by monocytes/macrophages, and promotes bone regeneration. However, the mechanisms involved in monocyte-Fg interaction, were only partially known. Current work addresses the interaction between primary human monocytes and Ch surfaces modified by Fg adsorption (Ch-Fg) at the molecular level. Results show that monocytes interact specifically with Ch-Fg via TLR-4, triggering particular intracellular signalling pathways (ERK and JNK, but not p38), downstream of TLR-4. Functionally, Ch-Fg induced monocytes to produce the osteogenic mediator BMP-2. Thus, we clarify herein two essential aspects of the interaction between adsorbed Fg and monocytes, with impact on immunomodulation and regeneration, upon biomaterial implantation.
Collapse
|
47
|
Bio-active molecules modified surfaces enhanced mesenchymal stem cell adhesion and proliferation. Biochem Biophys Res Commun 2016; 483:312-317. [PMID: 28025144 DOI: 10.1016/j.bbrc.2016.12.146] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 12/21/2016] [Indexed: 11/23/2022]
Abstract
Surface modification of the substrate as a component of in vitro cell culture and tissue engineering, using bio-active molecules including extracellular matrix (ECM) proteins or peptides derived ECM proteins can modulate the surface properties and thereby induce the desired signaling pathways in cells. The aim of this study was to evaluate the behavior of human bone marrow mesenchymal stem cells (hBM-MSCs) on glass substrates modified with fibronectin (Fn), collagen (Coll), RGD peptides (RGD) and designed peptide (R-pept) as bio-active molecules. The glass coverslips were coated with fibronectin, collagen, RGD peptide and R-peptide. Bone marrow mesenchymal stem cells were cultured on different substrates and the adhesion behavior in early incubation times was investigated using scanning electron microscopy (SEM) and confocal microscopy. The MTT assay was performed to evaluate the effect of different bio-active molecules on MSCs proliferation rate during 24 and 72 h. Formation of filopodia and focal adhesion (FA) complexes, two steps of cell adhesion process, were observed in MSCs cultured on bio-active molecules modified coverslips, specifically in Fn coated and R-pept coated groups. SEM image showed well adhesion pattern for MSCs cultured on Fn and R-pept after 2 h incubation, while the shape of cells cultured on Coll and RGD substrates indicated that they might experience stress condition in early hours of culture. Investigation of adhesion behavior, as well as proliferation pattern, suggests R-peptide as a promising bio-active molecule to be used for surface modification of substrate in supporting and inducing cell adhesion and proliferation.
Collapse
|
48
|
Clements LE, Garvican ER, Dudhia J, Smith RKW. Modulation of mesenchymal stem cell genotype and phenotype by extracellular matrix proteins. Connect Tissue Res 2016; 57:443-453. [PMID: 27448620 DOI: 10.1080/03008207.2016.1215442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AIM To investigate the effect of extracellular matrix (ECM) proteins on characteristics of mesenchymal stem cells (MSCs) and tendon-derived cells (TDCs). MATERIALS AND METHODS MSCs and TDCs, cultured in a monolayer (2D) or hydrogels (3D), with or without ECM protein supplementation, and on a non-viable native tendon (NNT) matrix were assayed for adhesion, proliferation, gene expression, and integrin expression. RESULTS MSCs exhibited a fibroblastic, spindle-shaped morphology on 2D matrices except in the presence of fibronectin. In 3D matrices, MSCs displayed a rounded phenotype except when cultured on NNTs where cells aligned along the collagen fibrils but, unlike TDCs, did not form inter-cellular cytoplasmic processes. MSC proliferation was significantly (p < 0.01) increased by collagen type I in 2D culture and fibronectin in 3D culture. TDC proliferation was unaffected by substrata. MSCs and TDCs differentially expressed α2 integrin. Adhesion to substrata was reduced by RGD-blocking peptide and β1 integrin antibody. The presence of collagen I or fibronectin upregulated MSC expression of collagen type I and collagen type III, COMP, decorin, osteopontin, and fibronectin. CONCLUSIONS The morphology, gene expression, and adhesion of both MSCs and TDCs are sensitive to the presence of specific ECM components. Interaction with the ECM is, therefore, likely to affect the mechanism of action of MSCs in vitro and may contribute to phenotypic modulation in vivo.
Collapse
Affiliation(s)
- Lucy E Clements
- a Department Clinical Sciences and Services , Royal Veterinary College , Hatfield , UK
| | - Elaine R Garvican
- a Department Clinical Sciences and Services , Royal Veterinary College , Hatfield , UK
| | - Jayesh Dudhia
- a Department Clinical Sciences and Services , Royal Veterinary College , Hatfield , UK
| | - Roger K W Smith
- a Department Clinical Sciences and Services , Royal Veterinary College , Hatfield , UK
| |
Collapse
|
49
|
Murgia A, Veronesi E, Candini O, Caselli A, D’souza N, Rasini V, Giorgini A, Catani F, Iughetti L, Dominici M, Burns JS. Potency Biomarker Signature Genes from Multiparametric Osteogenesis Assays: Will cGMP Human Bone Marrow Mesenchymal Stromal Cells Make Bone? PLoS One 2016; 11:e0163629. [PMID: 27711115 PMCID: PMC5053614 DOI: 10.1371/journal.pone.0163629] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 09/12/2016] [Indexed: 01/15/2023] Open
Abstract
In skeletal regeneration approaches using human bone marrow derived mesenchymal stromal cells (hBM-MSC), functional evaluation before implantation has traditionally used biomarkers identified using fetal bovine serum-based osteogenic induction media and time courses of at least two weeks. However, emerging pre-clinical evidence indicates donor-dependent discrepancies between these ex vivo measurements and the ability to form bone, calling for improved tests. Therefore, we adopted a multiparametric approach aiming to generate an osteogenic potency assay with improved correlation. hBM-MSC populations from six donors, each expanded under clinical-grade (cGMP) conditions, showed heterogeneity for ex vivo growth response, mineralization and bone-forming ability in a murine xenograft assay. A subset of literature-based biomarker genes was reproducibly upregulated to a significant extent across all populations as cells responded to two different osteogenic induction media. These 12 biomarkers were also measurable in a one-week assay, befitting clinical cell expansion time frames and cGMP growth conditions. They were selected for further challenge using a combinatorial approach aimed at determining ex vivo and in vivo consistency. We identified five globally relevant osteogenic signature genes, notably TGF-ß1 pathway interactors; ALPL, COL1A2, DCN, ELN and RUNX2. Used in agglomerative cluster analysis, they correctly grouped the bone-forming cell populations as distinct. Although donor #6 cells were correlation slope outliers, they contrastingly formed bone without showing ex vivo mineralization. Mathematical expression level normalization of the most discrepantly upregulated signature gene COL1A2, sufficed to cluster donor #6 with the bone-forming classification. Moreover, attenuating factors causing genuine COL1A2 gene down-regulation, restored ex vivo mineralization. This suggested that the signature gene had an osteogenically influential role; nonetheless no single biomarker was fully deterministic whereas all five signature genes together led to accurate cluster analysis. We show proof of principle for an osteogenic potency assay providing early characterization of primary cGMP-hBM-MSC cultures according to their donor-specific bone-forming potential.
Collapse
Affiliation(s)
- Alba Murgia
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Elena Veronesi
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
- TPM, Science & Technology Park for Medicine, Mirandola, Modena, Italia
| | - Olivia Candini
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Anna Caselli
- CVBF - Consorzio per le Valutazioni Biologiche e Farmacologiche, Ospedale Pediatrico Giovanni XXIII, Bari, Italia
| | - Naomi D’souza
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Valeria Rasini
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Andrea Giorgini
- Department of Orthopedic Surgery, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Fabio Catani
- Department of Orthopedic Surgery, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Lorenzo Iughetti
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
- TPM, Science & Technology Park for Medicine, Mirandola, Modena, Italia
- * E-mail: (MD); (JSB)
| | - Jorge S. Burns
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
- TPM, Science & Technology Park for Medicine, Mirandola, Modena, Italia
- * E-mail: (MD); (JSB)
| |
Collapse
|
50
|
Vilardell AM, Cinca N, Jokinen A, Garcia-Giralt N, Dosta S, Cano IG, Guilemany JM. Real-Time Protein and Cell Binding Measurements on Hydroxyapatite Coatings. J Funct Biomater 2016; 7:E23. [PMID: 27618911 PMCID: PMC5040996 DOI: 10.3390/jfb7030023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/27/2016] [Accepted: 08/16/2016] [Indexed: 12/31/2022] Open
Abstract
Although a lot of in vitro and in vivo assays have been performed during the last few decades years for hydroxyapatite bioactive coatings, there is a lack of exploitation of real-time in vitro interaction measurements. In the present work, real-time interactions for a plasma sprayed hydroxyapatite coating were measured by a Multi-Parametric Surface Plasmon Resonance (MP-SPR), and the results were compared with standard traditional cell viability in vitro assays. MP-SPR is proven to be suitable not only for measurement of molecule-molecule interactions but also molecule-material interaction measurements and cell interaction. Although SPR is extensively utilized in interaction studies, recent research of protein or cell adsorption on hydroxyapatite coatings for prostheses applications was not found. The as-sprayed hydroxyapatite coating resulted in 62.4% of crystalline phase and an average thickness of 24 ± 6 μm. The MP-SPR was used to measure lysozyme protein and human mesenchymal stem cells interaction to the hydroxyapatite coating. A comparison between the standard gold sensor and Hydroxyapatite (HA)-plasma coated sensor denoted a clearly favourable cell attachment on HA coated sensor as a significantly higher signal of cell binding was detected. Moreover, traditional cell viability and proliferation tests showed increased activity with culture time indicating that cells were proliferating on HA coating. Cells show homogeneous distribution and proliferation along the HA surface between one and seven days with no significant mortality. Cells were flattened and spread on rough surfaces from the first day, with increasing cytoplasmatic extensions during the culture time.
Collapse
Affiliation(s)
- A M Vilardell
- Centre de Projecció Tèrmica (CPT), Department Ciència dels Materials i Enginyeria Metal lúrgica, Universitat de Barcelona Martí i Franquès 1, Barcelona E-08028, Spain.
| | - N Cinca
- Centre de Projecció Tèrmica (CPT), Department Ciència dels Materials i Enginyeria Metal lúrgica, Universitat de Barcelona Martí i Franquès 1, Barcelona E-08028, Spain.
| | - A Jokinen
- BioNavis Ltd., Hermiankatu 6-8H, 33720 Tampere , Finland.
| | - N Garcia-Giralt
- URFOA, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), RETICEF, Doctor Aiguader 80, Barcelona 08003, Spain.
| | - S Dosta
- Centre de Projecció Tèrmica (CPT), Department Ciència dels Materials i Enginyeria Metal lúrgica, Universitat de Barcelona Martí i Franquès 1, Barcelona E-08028, Spain.
| | - I G Cano
- Centre de Projecció Tèrmica (CPT), Department Ciència dels Materials i Enginyeria Metal lúrgica, Universitat de Barcelona Martí i Franquès 1, Barcelona E-08028, Spain.
| | - J M Guilemany
- Centre de Projecció Tèrmica (CPT), Department Ciència dels Materials i Enginyeria Metal lúrgica, Universitat de Barcelona Martí i Franquès 1, Barcelona E-08028, Spain.
| |
Collapse
|