101
|
The Kinesin Gene KIF26B Modulates the Severity of Post-Traumatic Heterotopic Ossification. Int J Mol Sci 2022; 23:ijms23169203. [PMID: 36012474 PMCID: PMC9409126 DOI: 10.3390/ijms23169203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 11/17/2022] Open
Abstract
The formation of pathological bone deposits within soft tissues, termed heterotopic ossification (HO), is common after trauma. However, the severity of HO formation varies substantially between individuals, from relatively isolated small bone islands through to extensive soft tissue replacement by bone giving rise to debilitating symptoms. The aim of this study was to identify novel candidate therapeutic molecular targets for severe HO. We conducted a genome-wide scan in men and women with HO of varying severity following hip replacement for osteoarthritis. HO severity was dichotomized as mild or severe, and association analysis was performed with adjustment for age and sex. We next confirmed expression of the gene encoded by the lead signal in human bone and in primary human mesenchymal stem cells. We then examined the effect of gene knockout in a murine model of osseous trans-differentiation, and finally we explored transcription factor phosphorylation in key pathways perturbed by the gene. Ten independent signals were suggestively associated with HO severity, with KIF26B as the lead. We subsequently confirmed KIF26B expression in human bone and upregulation upon BMP2-induced osteogenic differentiation in primary human mesenchymal stem cells, and also in a rat tendo-Achilles model of post-traumatic HO. CRISPR-Cas9 mediated knockout of Kif26b inhibited BMP2-induced Runx2, Sp7/Osterix, Col1A1, Alp, and Bglap/Osteocalcin expression and mineralized nodule formation in a murine myocyte model of osteogenic trans-differentiation. Finally, KIF26B deficiency inhibited ERK MAP kinase activation during osteogenesis, whilst augmenting p38 and SMAD 1/5/8 phosphorylation. Taken together, these data suggest a role for KIF26B in modulating the severity of post-traumatic HO and provide a potential novel avenue for therapeutic translation.
Collapse
|
102
|
Chechekhin VI, Kulebyakin KY, Kokaev RI, Tyurin-Kuzmin PA. GPCRs in the regulation of the functional activity of multipotent mesenchymal stromal cells. Front Cell Dev Biol 2022; 10:953374. [PMID: 36046341 PMCID: PMC9421028 DOI: 10.3389/fcell.2022.953374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/01/2022] [Indexed: 11/24/2022] Open
Abstract
Adipose tissue is one of the tissues in the human body that is renewed during the whole life. Dysregulation of this process leads to conditions such as obesity, metabolic syndrome, and type 2 diabetes. The key role in maintaining the healthy state of adipose tissue is played by a specific group of postnatal stem cells called multipotent mesenchymal stromal cells (MSCs). They are both precursors for new adipocytes and key paracrine regulators of adipose tissue homeostasis. The activity of MSCs is tightly adjusted to the needs of the organism. To ensure such coordination, MSCs are put under strict regulation which is realized through a wide variety of signaling mechanisms. They control aspects of MSC activity such as proliferation, differentiation, and production of signal molecules via alteration of MSC sensitivity to hormonal stimuli. In this regard, MSCs use all the main mechanisms of hormonal sensitivity regulation observed in differentiated cells, but at the same time, several unique regulatory mechanisms have been found in MSCs. In the presented review, we will cover these unique mechanisms as well as specifics of common mechanisms of regulation of hormonal sensitivity in stem cells.
Collapse
Affiliation(s)
- Vadim I. Chechekhin
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Konstantin Yu. Kulebyakin
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Romesh I. Kokaev
- Institute of Biomedical Investigations, The Affiliate of Vladikavkaz Scientific Centre of Russian Academy of Sciences, Vladikavkaz, Russia
| | - Pyotr A. Tyurin-Kuzmin
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- *Correspondence: Pyotr A. Tyurin-Kuzmin,
| |
Collapse
|
103
|
Luo J, Yang L, Chueng STD, Conley B, Rathnam C, Lee KB. Advanced Drug Delivery Modulation via Hybrid Nanofibers Enhances Stem Cell Differentiation. ACS APPLIED MATERIALS & INTERFACES 2022; 14:34488-34501. [PMID: 35862271 PMCID: PMC9357201 DOI: 10.1021/acsami.2c10288] [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] [Indexed: 06/15/2023]
Abstract
Seamlessly integrating soluble factors onto biomedical scaffolds with a precisely manufactured topography for efficient cell control remains elusive since many scaffold fabrication techniques degrade payloads. Surface adsorption of payloads onto synthesized nanoscaffolds retains bioactivity by removing exposure to harsh processing conditions at the expense of inefficient drug loading and uncontrolled release. Herein, we present a nanomaterial composite scaffold paradigm to improve physicochemical surface adsorption pharmacokinetics. As a proof of concept, we integrated graphene oxide (GO) and manganese dioxide (MnO2) nanosheets onto nanofibers to increase loading capacity and tune drug release. Non-degradable GO enhances payload retention, while biodegradable MnO2 enables cell-responsive drug release. To demonstrate the utility of this hybrid nanomaterial scaffold paradigm for tissue engineering, we adsorbed payloads ranging from small molecules to proteins onto the scaffold to induce myogenesis and osteogenesis for multiple stem cell lines. Scaffolds with adsorbed payloads enabled more efficient differentiation than media supplementation using equivalent quantities of differentiation factors. We attribute this increased efficacy to a reverse uptake mechanism whereby payloads are localized around seeded cells, increasing delivery efficiency for guiding differentiation. Additionally, we demonstrate spatial control over cells since differentiation factors are delivered locally through the scaffold. When co-culturing scaffolds with and without adsorbed payloads, only cells seeded on payload-adsorbed scaffolds underwent differentiation. With this modular technology being capable of enhancing multiple differentiation fates for specific cell lines, this technology provides a promising alternative for current tissue engineering scaffolds.
Collapse
Affiliation(s)
- Jeffrey Luo
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Sy-Tsong Dean Chueng
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Brian Conley
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Christopher Rathnam
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
104
|
Azari Matin A, Fattah K, Saeidpour Masouleh S, Tavakoli R, Houshmandkia SA, Moliani A, Moghimimonfared R, Pakzad S, Dalir Abdolahinia E. Synthetic electrospun nanofibers as a supportive matrix in osteogenic differentiation of induced pluripotent stem cells. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:1469-1493. [PMID: 35321624 DOI: 10.1080/09205063.2022.2056941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Continuous remodeling is not able to repair large bone defects. Bone tissue engineering is aimed to repair these defects by creating bone grafts. To do this, several technologies and biomaterials have been employed to fabricate an in vivo-like supportive matrix. Electrospinning is a versatile technique to fabricate porous matrices with interconnected pores and high surface area, replicating in vivo microenvironment. Electrospun scaffolds have been used in a large number of studies to provide a matrix for bone regeneration and osteogenic differentiation of stem cells such as induced pluripotent stem cells (iPSCs). Electrospinning uses both natural and synthetic polymers, either alone or in combination, to fabricate scaffolds. Among them, synthetic polymers have had a great promise in bone regeneration and repair. They allow the fabrication of biocompatible and biodegradable scaffolds with high mechanical properties, suitable for bone engineering. Furthermore, several attempts have done to increase the osteogenic properties of these scaffolds. This paper reviewed the potential of synthetic electrospun scaffolds in osteogenic differentiation of iPSCs. In addition, the approaches to improve the osteogenic differentiation of these scaffolds are addressed.
Collapse
Affiliation(s)
- Arash Azari Matin
- Department of Biology, California State University, Northridge, CA, USA
| | - Khashayar Fattah
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Reza Tavakoli
- Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Afshin Moliani
- Isfahan Medical Students Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Moghimimonfared
- Department of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Sahar Pakzad
- Department of Oral and Maxillofacial Surgery, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elaheh Dalir Abdolahinia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
105
|
Kuo CH, Lee IC, Huang BJ, Chen CM, Liou YM. Effects of Aldo-Keto Reductase Family 1 Member A on Osteoblast Differentiation Associated with Lactate Production in MC3T3-E1 Preosteoblastic Cells. Biochem Cell Biol 2022; 100:413-424. [PMID: 35858481 DOI: 10.1139/bcb-2022-0108] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aldo-keto reductase family 1 member A (AKR1A) is an NADPH-dependent aldehyde reductase widely expressed in mammalian tissues. In this study, induced differentiation of MC3T3-E1 preosteoblasts was found to increase AKR1A gene expression concomitantly increased NOx- (nitrite+nitrate), increased glucose uptake, increased [NAD(P)+]/[NAD(P)H] and lactate production but decreased reactive oxygen species (ROS) without changes in eNOS (endothelial nitric oxide synthase) expression in differentiated osteoblasts (OBs). A study using gain- and loss-of-function MC3T3-E1 cells indicated that AKR1A is essential for modulating OB differentiation and gene expression of collagen1 A1, receptor activator of nuclear factor kappa-Β ligand, and osteoprotegerin in OBs. Immunofluorescence microscopy also revealed that changes in AKR1A expression altered extracellular collagen formation in differentiated OBs. Consistently, analyses of alkaline phosphatase activity and calcium deposits of matrix mineralization by Alizarin Red S staining verified that AKR1A is involved in the regulation of OB differentiation and bone matrix formation. In addition, AKR1A gene alterations affected the levels of NOx-, eNOS expression, glucose uptake, [NAD(P)+]/[NAD(P)H] dinucleotide redox couples, lactate production and ROS in differentiated OBs. Herein, we report that AKR1A-mediated denitrosylation may play a role in the regulation of lactate metabolism as well as redox homeostasis in cells, providing an efficient way to quickly gain energy and to significantly reduce oxidative stress for OB differentiation.
Collapse
Affiliation(s)
- Chia-Hsiao Kuo
- Tungs' Taichung MetroHarbor Hospital, 59084, Department of Orthopedics, Taichung, Taiwan;
| | - Inn-Chi Lee
- Chung Shan Medical University Taiyuan Road Branch, 63276, Taichung, Taiwan;
| | - Bo-Jun Huang
- National Chung Hsing University, 34916, LIFE SCIENCES, Taichung, Taiwan;
| | - Chuan-Mu Chen
- National Chung Hsing University, Department of Life Sciences, Taichung, Alberta, Taiwan;
| | - Ying-Ming Liou
- National Chung Hsing University, 34916, LIFE SCIENCES, Taichung, Please select an option below, Taiwan.,National Chung Hsing University, 34916, Rong Hsing Research Center for Translational Medicine, Taichung, Taiwan.,National Chung Hsing University, 34916, The iEGG and Animal Biotechnology Center, Taichung, Taiwan;
| |
Collapse
|
106
|
Li J, Hou W, Yang Y, Deng Q, Fu H, Yin Y, Duan K, Feng B, Guo T, Weng J. Micro/nano-topography promotes osteogenic differentiation of bone marrow stem cells by regulating periostin expression. Colloids Surf B Biointerfaces 2022; 218:112700. [PMID: 35907353 DOI: 10.1016/j.colsurfb.2022.112700] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 06/07/2022] [Accepted: 07/11/2022] [Indexed: 12/18/2022]
Abstract
Micro/nano-topography (MNT) is an important factor affecting cell response. Earlier studies using titania (TiO2) nanotube as a model of MNT found that they mediated the differentiation of BMSCs into osteoblasts, but the mechanisms are not fully understood. Surprisingly, Periostin (Postn), a secreted protein involved in extracellular matrix (ECM) construction and promoting osteogenic differentiation of bone marrow stem cells (BMSCs), was previously observed to significantly up-regulated on TiO2 nanotube. We proposed that Postn may act as a MNT signal transduction role. In this study, we investigated the effect of MNT on Postn, and the influence of Postn on osteogenic differentiation-related genes through focal adhesion and downstream signals. It was found that, titanium (Ti) plates carrying TiO2 nanotubes with diameters of ∼100 nm (TNT-100) significantly up-regulated the expression of Postn compared with flat Ti. Furthermore, Postn activated the downstream focal adhesion kinase (FAK) signal pathway and β-catenin into the nucleus by interacting with integrin αV. Surprisingly, TNT-100 up-regulated the transcription level of Wnt3a, which was independent of the up-regulation of Postn. This new Postn signaling pathway may provide more insights into the signal transduction mechanism of MNT and development of biomaterials with improved osteogenic properties.
Collapse
Affiliation(s)
- Jinsheng Li
- School of Materials Science & Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenqing Hou
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Yali Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Qing Deng
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Hong Fu
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Yiran Yin
- Sichuan Provincial Lab of Orthopaedic Engineering, Department of Bone and Joint Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Ke Duan
- Sichuan Provincial Lab of Orthopaedic Engineering, Department of Bone and Joint Surgery, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Bo Feng
- School of Materials Science & Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Tailin Guo
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| | - Jie Weng
- School of Materials Science & Engineering, Southwest Jiaotong University, Chengdu 610031, China; College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
107
|
Győrgy R, Kostoglou M, Mantalaris A, Georgiadis MC. Development of a multi-scale model to simulate Mesenchymal Stem Cell osteogenic differentiation within hydrogels in a rotating wall bioreactor. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2022.108566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
108
|
The Biological Basis for Surface-dependent Regulation of Osteogenesis and Implant Osseointegration. J Am Acad Orthop Surg 2022; 30:e894-e898. [PMID: 35383608 PMCID: PMC9464448 DOI: 10.5435/jaaos-d-21-00523] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 02/24/2022] [Indexed: 02/01/2023] Open
Abstract
Bone marrow stromal cells are regulated by the chemical and physical features of a biomaterial surface. When grown on titanium (Ti) and Ti alloy surfaces, such as titanium-aluminum-vanadium, with specific topographies that mimic the microscale, mesoscale, and nanoscale features of an osteoclast resorption pit, they undergo a rapid change in cell shape to assume a columnar morphology typical of a secretory osteoblast. These cells exhibit markers associated with an osteoblast phenotype, including osteocalcin and osteopontin, and they secrete factors associated with osteogenesis, including bone morphogenetic protein 2, vascular endothelial growth factor, and neurotrophic semaphorins. The pathway involves a shift in integrin expression from α5β1 to α2β1 and signaling by Wnt5a rather than Wnt3a. Conditioned media from these cultures can stimulate vasculogenesis by human endothelial cells and osteoblastic differentiation of marrow stromal cells not grown on the biomimetic substrate, suggesting that the surface could promote osteogenesis in vivo through similar mechanisms. In vivo studies using a variety of animal models confirm that implants with biomimetic surfaces result in improved osseointegration compared with Ti implants with smooth surfaces, as do meta-analyses comparing clinical performance of implant surface topographies.
Collapse
|
109
|
Humphreys PA, Mancini FE, Ferreira MJS, Woods S, Ogene L, Kimber SJ. Developmental principles informing human pluripotent stem cell differentiation to cartilage and bone. Semin Cell Dev Biol 2022; 127:17-36. [PMID: 34949507 DOI: 10.1016/j.semcdb.2021.11.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022]
Abstract
Human pluripotent stem cells can differentiate into any cell type given appropriate signals and hence have been used to research early human development of many tissues and diseases. Here, we review the major biological factors that regulate cartilage and bone development through the three main routes of neural crest, lateral plate mesoderm and paraxial mesoderm. We examine how these routes have been used in differentiation protocols that replicate skeletal development using human pluripotent stem cells and how these methods have been refined and improved over time. Finally, we discuss how pluripotent stem cells can be employed to understand human skeletal genetic diseases with a developmental origin and phenotype, and how developmental protocols have been applied to gain a better understanding of these conditions.
Collapse
Affiliation(s)
- Paul A Humphreys
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, University of Manchester, UK
| | - Fabrizio E Mancini
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Miguel J S Ferreira
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, University of Manchester, UK
| | - Steven Woods
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Leona Ogene
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| |
Collapse
|
110
|
Feier AM, Portan D, Manu DR, Kostopoulos V, Kotrotsos A, Strnad G, Dobreanu M, Salcudean A, Bataga T. Primary MSCs for Personalized Medicine: Ethical Challenges, Isolation and Biocompatibility Evaluation of 3D Electrospun and Printed Scaffolds. Biomedicines 2022; 10:biomedicines10071563. [PMID: 35884868 PMCID: PMC9313419 DOI: 10.3390/biomedicines10071563] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022] Open
Abstract
Autologous cell therapy uses patients’ own cells to deliver precise and ideal treatment through a personalized medicine approach. Isolation of patients’ cells from residual tissue extracted during surgery involves specific planning and lab steps. In the present manuscript, a path from isolation to in vitro research with human mesenchymal stem cells (MSCs) obtained from residual bone tissues is described as performed by a medical unit in collaboration with a research center. Ethical issues have been addressed by formulating appropriate harvesting protocols according to European regulations. Samples were collected from 19 patients; 10 of them were viable and after processing resulted in MSCs. MSCs were further differentiated in osteoblasts to investigate the biocompatibility of several 3D scaffolds produced by electrospinning and 3D printing technologies; traditional orthopedic titanium and nanostructured titanium substrates were also tested. 3D printed scaffolds proved superior compared to other substrates, enabling significantly improved response in osteoblast cells, indicating that their biomimetic structure and properties make them suitable for synthetic tissue engineering. The present research is a proof of concept that describes the process of primary stem cells isolation for in vitro research and opens avenues for the development of personalized cell platforms in the case of patients with orthopedic trauma. The demonstration model has promising perspectives in personalized medicine practices.
Collapse
Affiliation(s)
- Andrei Marian Feier
- Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Diana Portan
- Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (D.R.M.); (M.D.)
- Department of Mechanical Engineering and Aeronautics, University of Patras, Patras University Campus, 26504 Patras, Greece; (V.K.); (A.K.)
- Correspondence:
| | - Doina Ramona Manu
- Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (D.R.M.); (M.D.)
| | - Vassilis Kostopoulos
- Department of Mechanical Engineering and Aeronautics, University of Patras, Patras University Campus, 26504 Patras, Greece; (V.K.); (A.K.)
| | - Athanasios Kotrotsos
- Department of Mechanical Engineering and Aeronautics, University of Patras, Patras University Campus, 26504 Patras, Greece; (V.K.); (A.K.)
| | - Gabriela Strnad
- Faculty of Engineering and Information Technology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Minodora Dobreanu
- Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (D.R.M.); (M.D.)
| | - Andreea Salcudean
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Tiberiu Bataga
- Department of Orthopedics and Traumatology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| |
Collapse
|
111
|
Lin J, Sun M, Zhang J, Hu M, Zeng Y, Yi Q, Wang J, Bai Y, Zhang Y, Lu J. Solid‐state
NMR
(
SSNMR
) Characterization of Osteoblast from Mesenchymal Stromal Cell Differentiation to Osteoblast Mineralization. JBMR Plus 2022; 6:e10662. [PMID: 36248276 PMCID: PMC9549719 DOI: 10.1002/jbm4.10662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/25/2022] [Accepted: 06/09/2022] [Indexed: 11/19/2022] Open
Abstract
Solid‐state nuclear magnetic resonance (SSNMR), a technique capable of studying solid or semisolid biological samples, was first applied to study the cell differentiation and mineralization using the whole‐cell sample. Mesenchymal stromal cells (MSCs) with multipotent differentiation capacity were induced to differentiate into osteoblasts. The whole differentiation process, osteoblast mineralization and the mineral maturation, was investigated using SSNMR, providing intact, atomic level information on the cellular mineral structural transformation. Our research indicated the extent of osteoblast mineralization could vary significantly for different cell populations whereas the difference was not easily shown by other means of characterization. The SSNMR spectra revealed hydroxylapatite (or hydroxyapatite [HAP]) formation around 2 to 4 weeks after osteogenic induction for MSCs with a high differentiation potency. The early mineral phase deposit before HAP formation contained a high amount of HPO42−. The structures of minerals in the extracellular matrix (ECM) of osteoblasts could evolve for a period of time, even after the incubation of cells has been stopped. This observation was only possible by studying the sample in an intact state, where ECM was not disturbed. These findings improved our understanding of MSCs, which had wide applications in bone regeneration and tissue engineering. Meanwhile, this work demonstrated the advantage of studying these cellular systems as a whole without any mineral extraction, which had been largely overlooked. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jing‐Yu Lin
- School of Life Science and Technology ShanghaiTech University Shanghai China
- University of Chinese Academy of Sciences Beijing China
| | - Ming‐Hui Sun
- School of Life Science and Technology ShanghaiTech University Shanghai China
- University of Chinese Academy of Sciences Beijing China
| | - Jing Zhang
- School of Life Science and Technology ShanghaiTech University Shanghai China
- University of Chinese Academy of Sciences Beijing China
| | - Meng Hu
- School of Life Science and Technology ShanghaiTech University Shanghai China
- University of Chinese Academy of Sciences Beijing China
| | - Yu‐Teng Zeng
- School of Life Science and Technology ShanghaiTech University Shanghai China
- University of Chinese Academy of Sciences Beijing China
| | - Qian‐Qian Yi
- School of Life Science and Technology ShanghaiTech University Shanghai China
- University of Chinese Academy of Sciences Beijing China
| | - Jian Wang
- School of Life Science and Technology ShanghaiTech University Shanghai China
| | - Yun Bai
- School of Life Science and Technology ShanghaiTech University Shanghai China
| | - Yifeng Zhang
- School of Life Science and Technology ShanghaiTech University Shanghai China
- Shanghai Clinical Research and Trial Center ShanghaiTech University Shanghai China
| | - Jun‐Xia Lu
- School of Life Science and Technology ShanghaiTech University Shanghai China
| |
Collapse
|
112
|
Bloise N, Waldorff EI, Montagna G, Bruni G, Fassina L, Fang S, Zhang N, Jiang J, Ryaby JT, Visai L. Early Osteogenic Marker Expression in hMSCs Cultured onto Acid Etching-Derived Micro- and Nanotopography 3D-Printed Titanium Surfaces. Int J Mol Sci 2022; 23:7070. [PMID: 35806083 PMCID: PMC9266831 DOI: 10.3390/ijms23137070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 12/13/2022] Open
Abstract
Polyetheretherketone (PEEK) titanium composite (PTC) is a novel interbody fusion device that combines a PEEK core with titanium alloy (Ti6Al4V) endplates. The present study aimed to investigate the in vitro biological reactivity of human bone-marrow-derived mesenchymal stem cells (hBM-MSCs) to micro- and nanotopographies produced by an acid-etching process on the surface of 3D-printed PTC endplates. Optical profilometer and scanning electron microscopy were used to assess the surface roughness and identify the nano-features of etched or unetched PTC endplates, respectively. The viability, morphology and the expression of specific osteogenic markers were examined after 7 days of culture in the seeded cells. Haralick texture analysis was carried out on the unseeded endplates to correlate surface texture features to the biological data. The acid-etching process modified the surface roughness of the 3D-printed PTC endplates, creating micro- and nano-scale structures that significantly contributed to sustaining the viability of hBM-MSCs and triggering the expression of early osteogenic markers, such as alkaline phosphatase activity and bone-ECM protein production. Finally, the topography of 3D-printed PTC endplates influenced Haralick's features, which in turn correlated with the expression of two osteogenic markers, osteopontin and osteocalcin. Overall, these data demonstrate that the acid-etching process of PTC endplates created a favourable environment for osteogenic differentiation of hBM-MSCs and may potentially have clinical benefit.
Collapse
Affiliation(s)
- Nora Bloise
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, 27100 Pavia, Italy;
- Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100 Pavia, Italy
| | - Erik I. Waldorff
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Giulia Montagna
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, 27100 Pavia, Italy;
- Department of Electrical, Computer and Biomedical Engineering, Centre for Health Technologies (CHT), University of Pavia, 27100 Pavia, Italy;
| | - Giovanna Bruni
- C.S.G.I.-Department of Chemistry, Section of Physical Chemistry, University of Pavia, 27100 Pavia, Italy;
| | - Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering, Centre for Health Technologies (CHT), University of Pavia, 27100 Pavia, Italy;
| | - Samuel Fang
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Nianli Zhang
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Jiechao Jiang
- Department of Material Science, University of Texas, Arlington, TX 76019, USA;
| | - James T. Ryaby
- Research and Product Development, Orthofix Medical, Inc., 3451 Plano Parkway, Lewisville, TX 75056, USA; (E.I.W.); (S.F.); (N.Z.); (J.T.R.)
| | - Livia Visai
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, 27100 Pavia, Italy;
- Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100 Pavia, Italy
| |
Collapse
|
113
|
Bogdanova M, Zabirnyk A, Malashicheva A, Semenova D, Kvitting JPE, Kaljusto ML, Perez MDM, Kostareva A, Stensløkken KO, Sullivan GJ, Rutkovskiy A, Vaage J. Models and Techniques to Study Aortic Valve Calcification in Vitro, ex Vivo and in Vivo. An Overview. Front Pharmacol 2022; 13:835825. [PMID: 35721220 PMCID: PMC9203042 DOI: 10.3389/fphar.2022.835825] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Aortic valve stenosis secondary to aortic valve calcification is the most common valve disease in the Western world. Calcification is a result of pathological proliferation and osteogenic differentiation of resident valve interstitial cells. To develop non-surgical treatments, the molecular and cellular mechanisms of pathological calcification must be revealed. In the current overview, we present methods for evaluation of calcification in different ex vivo, in vitro and in vivo situations including imaging in patients. The latter include echocardiography, scanning with computed tomography and magnetic resonance imaging. Particular emphasis is on translational studies of calcific aortic valve stenosis with a special focus on cell culture using human primary cell cultures. Such models are widely used and suitable for screening of drugs against calcification. Animal models are presented, but there is no animal model that faithfully mimics human calcific aortic valve disease. A model of experimentally induced calcification in whole porcine aortic valve leaflets ex vivo is also included. Finally, miscellaneous methods and aspects of aortic valve calcification, such as, for instance, biomarkers are presented.
Collapse
Affiliation(s)
- Maria Bogdanova
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Arsenii Zabirnyk
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Anna Malashicheva
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Daria Semenova
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | | | - Mari-Liis Kaljusto
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | | | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Woman and Children Health, Karolinska Institute, Stockholm, Sweden
| | - Kåre-Olav Stensløkken
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Gareth J Sullivan
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Institute of Immunology, Oslo University Hospital, Oslo, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Arkady Rutkovskiy
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pulmonary Diseases, Oslo University Hospital, Oslo, Norway
| | - Jarle Vaage
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
114
|
Li SJ, Cheng WL, Kao YH, Chung CC, Trang NN, Chen YJ. Melatonin Inhibits NF-κB/CREB/Runx2 Signaling and Alleviates Aortic Valve Calcification. Front Cardiovasc Med 2022; 9:885293. [PMID: 35795373 PMCID: PMC9251177 DOI: 10.3389/fcvm.2022.885293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is linked to high mortality. Melatonin inhibits nuclear factor-kappa B (NF-κB)/cyclic AMP response element-binding protein (CREB), contributing to CAVD progression. This study determined the role of melatonin/MT1/MT2 signaling in valvular interstitial cell (VIC) calcification. Western blotting and Alizarin red staining were used to analyze NF-κB/CREB/runt-related transcription factor 2 (Runx2) signaling in porcine VICs treated with an osteogenic (OST) medium without (control) or with melatonin for 5 days. Chromatin immunoprecipitation (ChIP) assay was used to analyze NF-κB's transcription regulation of NF-κB on the Runx2 promoter. OST medium-treated VICs exhibited a greater expression of NF-κB, CREB, and Runx2 than control VICs. Melatonin treatment downregulated the effects of the OST medium and reduced VIC calcification. The MT1/MT2 antagonist (Luzindole) and MT1 receptor neutralized antibody blocked the anticalcification effect of melatonin, but an MT2-specific inhibitor (4-P-PDOT) did not. Besides, the NF-κB inhibitor (SC75741) reduced OST medium-induced VIC calcification to a similar extent to melatonin at 10 nmol/L. The ChIP assay demonstrated that melatonin attenuated OST media increased NF-κB binding activity to the promoter region of Runx2. Activation of the melatonin/MT1-axis significantly reduced VIC calcification by targeting the NF-κB/CREB/Runx2 pathway. Targeting melatonin/MT1 signaling may be a potential therapeutic strategy for CAVD.
Collapse
Affiliation(s)
- Shao-Jung Li
- Division of Cardiovascular Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Wan-Li Cheng
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Chih Chung
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | - Yi-Jen Chen
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- *Correspondence: Yi-Jen Chen
| |
Collapse
|
115
|
Perez JE, Bajaber B, Alsharif N, Martínez-Banderas AI, Patel N, Sharip A, Di Fabrizio E, Merzaban J, Kosel J. Modulated nanowire scaffold for highly efficient differentiation of mesenchymal stem cells. J Nanobiotechnology 2022; 20:282. [PMID: 35710420 PMCID: PMC9202102 DOI: 10.1186/s12951-022-01488-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/31/2022] [Indexed: 12/18/2022] Open
Abstract
Background Nanotopographical cues play a critical role as drivers of mesenchymal stem cell differentiation. Nanowire scaffolds, in this regard, provide unique and adaptable nanostructured surfaces with focal points for adhesion and with elastic properties determined by nanowire stiffness. Results We show that a scaffold of nanowires, which are remotely actuated by a magnetic field, mechanically stimulates mesenchymal stem cells. Osteopontin, a marker of osteogenesis onset, was expressed after cells were cultured for 1 week on top of the scaffold. Applying a magnetic field significantly boosted differentiation due to mechanical stimulation of the cells by the active deflection of the nanowire tips. The onset of differentiation was reduced to 2 days of culture based on the upregulation of several osteogenesis markers. Moreover, this was observed in the absence of any external differentiation factors. Conclusions The magneto-mechanically modulated nanosurface enhanced the osteogenic differentiation capabilities of mesenchymal stem cells, and it provides a customizable tool for stem cell research and tissue engineering. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01488-5.
Collapse
Affiliation(s)
- Jose E Perez
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Bashaer Bajaber
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Nouf Alsharif
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Aldo I Martínez-Banderas
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Niketan Patel
- Electrical and Computer Engineering Program, Computer, Electrical and Mathematical Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Ainur Sharip
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Enzo Di Fabrizio
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129, Turin, Italy
| | - Jasmeen Merzaban
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia.
| | - Jürgen Kosel
- Electrical and Computer Engineering Program, Computer, Electrical and Mathematical Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia. .,Division of Sensor Systems, Silicon Austria Labs, High Tech Campus Villach, 9524, Villach, Austria.
| |
Collapse
|
116
|
Gerdesmeyer L, Zielhardt P, Klüter T, Gollwitzer H, Gerdesmeyer L, Hausdorf J, Ringeisen M, Knobloch K, Saxena A, Krath A. Stimulation of human bone marrow mesenchymal stem cells by electromagnetic transduction therapy - EMTT. Electromagn Biol Med 2022; 41:304-314. [DOI: 10.1080/15368378.2022.2079672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Ludger Gerdesmeyer
- Department of Sports Medicine, Palo Alto Medical Center, Palo Alto, USA
- Department of Orthopedic and Traumatology, Technical University Munich, Munich Germany
- Städtisches Krankenhaus Kiel, Kiel
| | - Paula Zielhardt
- Department of Orthopaedic Surgery and Traumatology, University Schleswig Holstein, Kiel, Germany
| | - Tim Klüter
- Department of Orthopaedic Surgery and Traumatology, University Schleswig Holstein, Kiel, Germany
| | - Hans Gollwitzer
- Department of Orthopedic and Traumatology, Technical University Munich, Munich Germany
| | - Lennart Gerdesmeyer
- Department of Orthopedic and Traumatology, Technical University Munich, Munich Germany
| | - Joerg Hausdorf
- Orthopedic Department, Physical Medicine and Rehabilitation, University Hospital of Munich, Germany
| | - Martin Ringeisen
- Department of Orthopedic Surgery and Traumatology, Orthopaedic Medical Center Dr. Ringeisen, Augsburg, Germany
| | - Karsten Knobloch
- Orthopedic Department, SportPraxis Prof. Dr. med. Karsten Knobloch, Hannover
| | - Amol Saxena
- Department of Sports Medicine, Palo Alto Medical Center, Palo Alto, USA
| | - André Krath
- Department of Orthopaedic Surgery and Traumatology, University Schleswig Holstein, Kiel, Germany
| |
Collapse
|
117
|
Effects of Different Basal Cell Culture Media upon the Osteogenic Response of hMSCs Evaluated by 99mTc-HDP Labeling. Int J Mol Sci 2022; 23:ijms23116288. [PMID: 35682966 PMCID: PMC9181423 DOI: 10.3390/ijms23116288] [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: 03/25/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/04/2022] Open
Abstract
The osteogenic differentiation of mesenchymal stem cells is now a standard procedure in modern bone tissue engineering. As this is a promising field for future clinical applications, many cell culture media exist to promote osteogenic differentiation. Prior to differentiation, cells must be expanded to obtain sufficient numbers for experiments. Little evidence is available regarding the optimal media combination for expansion and differentiation to maximize the osteogenic response. Therefore, human BM-MSCs (n = 6) were expanded in parallel in DMEM (Dulbecco’s Modified Eagle Medium) LG (Low Glucose) and α-MEM (Minimum Essential Media alpha-modification), followed by simultaneous monolayer differentiation toward the osteogenic lineage in: 1. DMEM LG (Low Glucose), 2. DMEM HG (High Glucose), 3. α-MEM, 4. “Bernese medium”, and 5. “Verfaillie medium”, with a corresponding negative control (total 20 groups). As a marker for osteogenic differentiation, hydroxyapatite was accessed using radioactive 99mTc-HDP labeling and quantitative alizarin red staining. The results indicate that all media except “Bernese medium” are suitable for osteogenic differentiation, while there was evidence that DMEM LG is partly superior when used for expansion and differentiation of BM-hMSCs. Using “Verfaillie medium” after DMEM LG expansion led to the highest grade of osteogenic differentiation. Nevertheless, the difference was not significant. Therefore, we recommend using DMEM LG for robust osteogenic differentiation, as it is highly suitable for that purpose, economical compared to other media, and requires little preparation time.
Collapse
|
118
|
He J, You D, Li Q, Wang J, Ding S, He X, Zheng H, Ji Z, Wang X, Ye X, Liu C, Kang H, Xu X, Xu X, Wang H, Yu M. Osteogenesis-Inducing Chemical Cues Enhance the Mechanosensitivity of Human Mesenchymal Stem Cells for Osteogenic Differentiation on a Microtopographically Patterned Surface. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200053. [PMID: 35373921 PMCID: PMC9165486 DOI: 10.1002/advs.202200053] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/25/2022] [Indexed: 05/13/2023]
Abstract
Mechanical cues are widely used for regulating cell behavior because of their overarching, extensive, and non-invasive advantages. However, unlike chemical cues, mechanical cues are not efficient enough to determine cell fate independently and improving the mechanosensitivity of cells is rather challenging. In this study, the combined effect of chemical and mechanical cues on the osteogenic differentiation of human mesenchymal stem cells is examined. These results show that chemical cues such as the presence of an osteogenic medium, induce cells to secrete more collagen, and induce integrin for recruiting focal adhesion proteins that mature and cascade a series of events with the help of the mechanical force of the scaffold material. High-resolution, highly ordered hollow-micro-frustum-arrays using double-layer lithography, combined with modified methacrylate gelatin loaded with pre-defined soluble chemicals to provide both chemical and mechanical cues to cells. This approach ultimately facilitates the achievement of cellular osteodifferentiation and enhances bone repair efficiency in a model of femoral fracture in vivo in mice. Moreover, the results also reveal these pivotal roles of Integrin α2/Focal adhesion kinase/Ras homolog gene family member A/Large Tumor Suppressor 1/Yes-associated protein in human mesenchymal stem cells osteogenic differentiation both in vitro and in vivo. Overall, these results show that chemical cues enhance the microtopographical sensitivity of cells.
Collapse
Affiliation(s)
- Jianxiang He
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Dongqi You
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Qi Li
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Jiabao Wang
- School of Materials Science and Engineeringand Institute for Advanced StudyTongji UniversityShanghai201804P. R. China
| | - Sijia Ding
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Xiaotong He
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Haiyan Zheng
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Zhenkai Ji
- School of Materials Science and Engineeringand Institute for Advanced StudyTongji UniversityShanghai201804P. R. China
| | - Xia Wang
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Xin Ye
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Chao Liu
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| | - Hanyue Kang
- School of Materials Science and Engineeringand Institute for Advanced StudyTongji UniversityShanghai201804P. R. China
| | - Xiuzhen Xu
- School of Materials Science and Engineeringand Institute for Advanced StudyTongji UniversityShanghai201804P. R. China
| | - Xiaobin Xu
- School of Materials Science and Engineeringand Institute for Advanced StudyTongji UniversityShanghai201804P. R. China
| | - Huiming Wang
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
- School of StomatologyThe First Affiliated Hospital of Zhejiang University School of MedicineHangzhou310003P. R. China
| | - Mengfei Yu
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceStomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesHangzhou310006P. R. China
| |
Collapse
|
119
|
Li T, Geng Y, Hu Y, Zhang L, Cui X, Zhang W, Gao F, Liu Z, Luo X. Dentin Matrix Protein 1 Silencing Inhibits Phosphorus Utilization in Primary Cultured Tibial Osteoblasts of Broiler Chicks. Front Vet Sci 2022; 9:875140. [PMID: 35558889 PMCID: PMC9087580 DOI: 10.3389/fvets.2022.875140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 02/21/2022] [Indexed: 12/19/2022] Open
Abstract
Three experiments were carried out in the present study to investigate whether dentin matrix protein 1 (DMP1) was involved in regulating phosphorus (P) metabolic utilization in primary cultured tibial osteoblasts of broiler chicks. Experiment 1 was conducted to select the optimal osteogenic inductive culture medium and the optimal induction time in primary cultured tibial osteoblasts of broiler chicks. In experiment 2, the siRNAs against DMP1 were designed, synthesized and transfected into primary cultured tibial osteoblasts of broiler chicks, and then the inhibitory efficiencies of siRNAs against DMP1 were determined, and the most efficacious siRNA was selected to be used for the DMP1 silencing. In experiment 3, with or without siRNA against DMP1, primary cultured tibial osteoblasts of broiler chicks were treated with the medium supplemented with 0.0, 1.0 or 2.0 mmol/L of P as NaH2PO4 for 12 days. The P metabolic utilization-related parameters were measured. The results showed that the osteogenic induced medium 2 and 12 days of the optimal induction time were selected; Among the designed siRNAs, the si340 was the most effective (P < 0.05) in inhibiting the DMP1 expression; DMP1 silencing decreased (P < 0.05) the expressions of DMP1 mRNA and protein, P retention rate, mineralization formation, alkaline phosphatase activity and bone gla-protein content in tibial osteoblasts at all of added P levels. It is concluded that DMP1 silencing inhibited P utilization, and thus DMP1 was involved in regulating P metabolic utilization in primary cultured tibial osteoblasts of broiler chicks, which provides a novel insight into the regulation of the P utilization in the bone of broilers, and will contribute to develop feasible strategies to improve the bone P utilization efficiency of broilers so as to decrease its excretion.
Collapse
Affiliation(s)
- Tingting Li
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanqiang Geng
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yun Hu
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Liyang Zhang
- Mineral Nutrition Research Division, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoyan Cui
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Weiyun Zhang
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Feiyu Gao
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Xugang Luo
- Poultry Mineral Nutrition Laboratory, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- *Correspondence: Xugang Luo
| |
Collapse
|
120
|
Yadav LR, Balagangadharan K, Lavanya K, Selvamurugan N. Orsellinic acid-loaded chitosan nanoparticles in gelatin/nanohydroxyapatite scaffolds for bone formation in vitro. Life Sci 2022; 299:120559. [PMID: 35447131 DOI: 10.1016/j.lfs.2022.120559] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
AIM Orsellinic acid (2,4-Dimethoxy-6-methylbenzoic acid) (OA) is a hydrophobic polyphenolic compound with therapeutic potential, but its impact on actuating osteogenesis remains unknown. The bioavailability of OA is hampered by its hydrophobic nature. This study aimed to fabricate nano-drug delivery system-based scaffolds for OA and test its potential for osteogenesis in vitro. MATERIALS AND METHODS OA was loaded into chitosan nanoparticles (nCS + OA) using the ionic gelation technique at different concentrations. nCS + OA were incorporated onto the scaffolds containing gelatin (Gel) and nanohydroxyapatite (nHAp) by the lyophilization method. Biocomposite scaffolds were examined for their physicochemical and material characteristic properties. The effect of OA in the scaffolds for osteoblast differentiation was determined by alizarin red and von Kossa staining at the cellular level and by reverse transcriptase-qPCR and western blot analysis at the molecular level. KEY FINDINGS The scaffolds showed excellent physiochemical and material characteristics and remained cyto-friendly to mouse mesenchymal stem cells (mMSCs, C3H10T1/2). The release of OA from Gel/nHAp/nCS scaffolds enhanced the differentiation of mMSCs towards osteoblasts, as observed through cellular and molecular studies. Moreover, the osteogenic potential of OA was mediated by the activation of FAK and ERK signaling pathways through integrins. SIGNIFICANCE The inclusion of OA into Gel/nHAp/nCS biocomposite scaffolds at 80 μM concentration promoted osteoblast differentiation via cell adhesion mediated signaling, compared with that shown by Gel/nHAp/nCS alone. Overall, this study identified the potential therapeutic OA containing Gel/nHAp/nCS scaffolds, accelerating its potential for clinical application towards bone regeneration.
Collapse
Affiliation(s)
- L Roshini Yadav
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - K Balagangadharan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - K Lavanya
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
121
|
TBP, PPIA, YWHAZ and EF1A1 Are the Most Stably Expressed Genes during Osteogenic Differentiation. Int J Mol Sci 2022; 23:ijms23084257. [PMID: 35457075 PMCID: PMC9025278 DOI: 10.3390/ijms23084257] [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: 03/16/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
RT-qPCR is the gold standard and the most commonly used method for measuring gene expression. Selection of appropriate reference gene(s) for normalization is a crucial part of RT-qPCR experimental design, which allows accurate quantification and reliability of the results. Because there is no universal reference gene and even commonly used housekeeping genes’ expression can vary under certain conditions, careful selection of an appropriate internal control must be performed for each cell type or tissue and experimental design. The aim of this study was to identify the most stable reference genes during osteogenic differentiation of the human osteosarcoma cell lines MG-63, HOS, and SaOS-2 using the geNorm, NormFinder, and BestKeeper statistical algorithms. Our results show that TBP, PPIA, YWHAZ, and EF1A1 are the most stably expressed genes, while ACTB, and 18S rRNA expressions are most variable. These data provide a basis for future RT-qPCR normalizations when studying gene expression during osteogenic differentiation, for example, in studies of osteoporosis and other bone diseases.
Collapse
|
122
|
Endo- and Exometabolome Crosstalk in Mesenchymal Stem Cells Undergoing Osteogenic Differentiation. Cells 2022; 11:cells11081257. [PMID: 35455937 PMCID: PMC9024772 DOI: 10.3390/cells11081257] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
This paper describes, for the first time to our knowledge, a lipidome and exometabolome characterization of osteogenic differentiation for human adipose tissue stem cells (hAMSCs) using nuclear magnetic resonance (NMR) spectroscopy. The holistic nature of NMR enabled the time-course evolution of cholesterol, mono- and polyunsaturated fatty acids (including ω-6 and ω-3 fatty acids), several phospholipids (phosphatidylcholine, phosphatidylethanolamine, sphingomyelins, and plasmalogens), and mono- and triglycerides to be followed. Lipid changes occurred almost exclusively between days 1 and 7, followed by a tendency for lipidome stabilization after day 7. On average, phospholipids and longer and more unsaturated fatty acids increased up to day 7, probably related to plasma membrane fluidity. Articulation of lipidome changes with previously reported polar endometabolome profiling and with exometabolome changes reported here in the same cells, enabled important correlations to be established during hAMSC osteogenic differentiation. Our results supported hypotheses related to the dynamics of membrane remodelling, anti-oxidative mechanisms, protein synthesis, and energy metabolism. Importantly, the observation of specific up-taken or excreted metabolites paves the way for the identification of potential osteoinductive metabolites useful for optimized osteogenic protocols.
Collapse
|
123
|
Montorsi M, Genchi GG, De Pasquale D, De Simoni G, Sinibaldi E, Ciofani G. Design, Fabrication, and Characterization of a Multimodal Reconfigurable Bioreactor for Bone Tissue Engineering. Biotechnol Bioeng 2022; 119:1965-1979. [PMID: 35383894 PMCID: PMC9324218 DOI: 10.1002/bit.28100] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/15/2022] [Accepted: 03/31/2022] [Indexed: 11/18/2022]
Abstract
In the past decades, bone tissue engineering developed and exploited many typologies of bioreactors, which, besides providing proper culture conditions, aimed at integrating those bio‐physical stimulations that cells experience in vivo, to promote osteogenic differentiation. Nevertheless, the highly challenging combination and deployment of many stimulation systems into a single bioreactor led to the generation of several unimodal bioreactors, investigating one or at mostly two of the required biophysical stimuli. These systems miss the physiological mimicry of bone cells environment, and often produced contrasting results, thus making the knowledge of bone mechanotransduction fragmented and often inconsistent. To overcome this issue, in this study we developed a perfusion and electroactive‐vibrational reconfigurable stimulation bioreactor to investigate the differentiation of SaOS‐2 bone‐derived cells, hosting a piezoelectric nanocomposite membrane as cell culture substrate. This multimodal perfusion bioreactor is designed based on a numerical (finite element) model aimed at assessing the possibility to induce membrane nano‐scaled vibrations (with ~12 nm amplitude at a frequency of 939 kHz) during perfusion (featuring 1.46 dyn cm−2 wall shear stress), large enough for inducing a physiologically‐relevant electric output (in the order of 10 mV on average) on the membrane surface. This study explored the effects of different stimuli individually, enabling to switch on one stimulation at a time, and then to combine them to induce a faster bone matrix deposition rate. Biological results demonstrate that the multimodal configuration is the most effective in inducing SaOS‐2 cell differentiation, leading to 20‐fold higher collagen deposition compared to static cultures, and to 1.6‐ and 1.2‐fold higher deposition than the perfused‐ or vibrated‐only cultures. These promising results can provide tissue engineering scientists with a comprehensive and biomimetic stimulation platform for a better understanding of mechanotransduction phenomena beyond cells differentiation.
Collapse
Affiliation(s)
- Margherita Montorsi
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy.,Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| | - Giada Graziana Genchi
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| | - Daniele De Pasquale
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| | - Giorgio De Simoni
- CNR, Nanoscience Institute, NEST Laboratory, Piazza San Silvestro 12, 56127, Pisa, Italy
| | - Edoardo Sinibaldi
- Istituto Italiano di Tecnologia, Bioinspired Soft Robotics, Via Morego 30, 16163, Genova, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| |
Collapse
|
124
|
Impact of Treadmill Interval Running on the Appearance of Zinc Finger Protein FHL2 in Bone Marrow Cells in a Rat Model: A Pilot Study. Life (Basel) 2022; 12:life12040528. [PMID: 35455019 PMCID: PMC9029125 DOI: 10.3390/life12040528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Although the benefits of physical exercise to preserve bone quality are now widely recognized, the intimate mechanisms leading to the underlying cell responses still require further investigations. Interval training running, for instance, appears as a generator of impacts on the skeleton, and particularly on the progenitor cells located in the bone marrow. Therefore, if this kind of stimulus initiates bone cell proliferation and differentiation, the activation of a devoted signaling pathway by mechano-transduction seems likely. This study aimed at investigating the effects of an interval running program on the appearance of the zinc finger protein FHL2 in bone cells and their anatomical location. Twelve 5-week-old male Wistar rats were randomly allocated to one of the following groups (n = 6 per group): sedentary control (SED) or high-intensity interval running (EX, 8 consecutive weeks). FHL2 identification in bone cells was performed by immuno-histochemistry on serial sections of radii. We hypothesized that impacts generated by running could activate, in vivo, a specific signaling pathway, through an integrin-mediated mechano-transductive process, leading to the synthesis of FHL2 in bone marrow cells. Our data demonstrated the systematic appearance of FHL2 (% labeled cells: 7.5%, p < 0.001) in bone marrow obtained from EX rats, whereas no FHL2 was revealed in SED rats. These results suggest that the mechanical impacts generated during high-intensity interval running activate a signaling pathway involving nuclear FHL2, such as that also observed with dexamethasone administration. Consequently, interval running could be proposed as a non-pharmacological strategy to contribute to bone marrow cell osteogenic differentiation.
Collapse
|
125
|
Tanaka M, Izumiya M, Haniu H, Ueda K, Ma C, Ueshiba K, Ideta H, Sobajima A, Uchiyama S, Takahashi J, Saito N. Current Methods in the Study of Nanomaterials for Bone Regeneration. NANOMATERIALS 2022; 12:nano12071195. [PMID: 35407313 PMCID: PMC9000656 DOI: 10.3390/nano12071195] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 12/18/2022]
Abstract
Nanomaterials show great promise as bone regeneration materials. They can be used as fillers to strengthen bone regeneration scaffolds, or employed in their natural form as carriers for drug delivery systems. A variety of experiments have been conducted to evaluate the osteogenic potential of bone regeneration materials. In vivo, such materials are commonly tested in animal bone defect models to assess their bone regeneration potential. From an ethical standpoint, however, animal experiments should be minimized. A standardized in vitro strategy for this purpose is desirable, but at present, the results of studies conducted under a wide variety of conditions have all been evaluated equally. This review will first briefly introduce several bone regeneration reports on nanomaterials and the nanosize-derived caveats of evaluations in such studies. Then, experimental techniques (in vivo and in vitro), types of cells, culture media, fetal bovine serum, and additives will be described, with specific examples of the risks of various culture conditions leading to erroneous conclusions in biomaterial analysis. We hope that this review will create a better understanding of the evaluation of biomaterials, including nanomaterials for bone regeneration, and lead to the development of versatile assessment methods that can be widely used in biomaterial development.
Collapse
Affiliation(s)
- Manabu Tanaka
- Department of Orthopedic Surgery, Okaya City Hospital, 4-11-33 Honcho, Okaya, Nagano 394-8512, Japan;
- Correspondence: (M.T.); (H.H.); Tel.: +81-266-23-8000 (M.T.); +81-263-37-3555 (H.H.)
| | - Makoto Izumiya
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Hisao Haniu
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
- Correspondence: (M.T.); (H.H.); Tel.: +81-266-23-8000 (M.T.); +81-263-37-3555 (H.H.)
| | - Katsuya Ueda
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Chuang Ma
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Koki Ueshiba
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Hirokazu Ideta
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
| | - Atsushi Sobajima
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
- Department of Orthopedics (Lower Limbs), Social Medical Care Corporation Hosei-kai Marunouchi Hospital, 1-7-45 Nagisa, Matsumoto, Nagano 390-8601, Japan
| | - Shigeharu Uchiyama
- Department of Orthopedic Surgery, Okaya City Hospital, 4-11-33 Honcho, Okaya, Nagano 394-8512, Japan;
| | - Jun Takahashi
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
| | - Naoto Saito
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
| |
Collapse
|
126
|
Ricci A, Cataldi A, Zara S, Gallorini M. Graphene-Oxide-Enriched Biomaterials: A Focus on Osteo and Chondroinductive Properties and Immunomodulation. MATERIALS (BASEL, SWITZERLAND) 2022; 15:2229. [PMID: 35329679 PMCID: PMC8955105 DOI: 10.3390/ma15062229] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/28/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022]
Abstract
Due to its exceptional physical properties, such as high electronic conductivity, good thermal stability, excellent mechanical strength, and chemical versatility, graphene has sparked a lot of interest in the scientific community for various applications. It has therefore been employed as an antibacterial agent, in photothermal therapy (PTT) and biosensors, in gene delivery systems, and in tissue engineering for regenerative purposes. Since it was first discovered in 1947, different graphene derivatives have been synthetized from pristine graphene. The most adaptable derivate is graphene oxide (GO). Owing to different functional groups, the amphiphilic structure of GO can interact with cells and exogenous or endogenous growth/differentiation factors, allowing cell adhesion, growth, and differentiation. When GO is used as a coating for scaffolds and nanomaterials, it has been found to enhance bone, chondrogenic, cardiac, neuronal, and skin regeneration. This review focuses on the applications of graphene-based materials, in particular GO, as a coating for scaffolds in bone and chondrogenic tissue engineering and summarizes the most recent findings. Moreover, novel developments on the immunomodulatory properties of GO are reported.
Collapse
Affiliation(s)
| | | | | | - Marialucia Gallorini
- Department of Pharmacy, “G. d'Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (A.R.); (A.C.); (S.Z.)
| |
Collapse
|
127
|
Ahmadi F, Shaidi S, Hadipour E, Khodaverdi E, Hadizadeh F, Kamali H, Tayarani-Najaran Z. Effects of Dexamethasone-In Situ Forming Implant (ISFI) on the Differentiation Process of Human Dental Pulp Stem Cells to Osteoblasts. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00255-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
128
|
Bordini EAF, Cassiano FB, Bronze-Uhle ES, Alamo L, Hebling J, de Souza Costa CA, Soares DG. Chitosan in association with osteogenic factors as a cell-homing platform for dentin regeneration: Analysis in a pulp-in-a-chip model. Dent Mater 2022; 38:655-669. [PMID: 35210124 DOI: 10.1016/j.dental.2022.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 01/04/2022] [Accepted: 02/12/2022] [Indexed: 12/18/2022]
Abstract
OBJECTIVE In this paper we propose the association of β-glycerophosphate (βGP) and calcium-hydroxide with chitosan (CH) to formulate a porous bioactive scaffold suitable as a cell-homing platform for dentin regeneration. METHODS Calcium hydroxide and βGP solutions were incorporated into chitosan to modulate scaffold architecture and composition by a phase separation technique. Architecture, chemical composition, and degradability were evaluated, and biological characterizations were performed by the seeding of dental pulp cells (DPCs) onto scaffolds, or by cultivating them in contact with leachable components (extracts), to determine cytocompatibility and odontoblastic differentiation. Cell-free scaffolds were then positioned in intimate contact with a 3D culture of DPCs in a pulp-in-a-chip platform under simulated pulp pressure. Cell mobilization and odontoblastic marker expression were evaluated. Deposition of mineralized matrix was assessed in direct contact with dentin, in the absence of osteogenic factors. RESULTS Incorporation of calcium hydroxide and βGP generated a stable porous chitosan scaffold containing Ca-P nanoglobule topography (CH-Ca-βGP), which favored cell viability, alkaline phosphatase activity, and mineralized matrix deposition by cells seeded onto the scaffold structure and at a distance. The pulp-in-a-chip assay denoted its chemotactic and bioactive potential, since dentin sialoprotein-positive DPCs from 3D culture adhered to CH-Ca-βGP more than to plain chitosan. The higher deposition of mineralized matrix onto the scaffold and surrounding dentin was also observed. SIGNIFICANCE A CH-Ca-βGP scaffold creates a microenvironment capable of mobilizing DPC migration toward its structure, harnessing the odontogenic potential and culminating in the expression of a highly mineralizing phenotype, key factors for a cell-homing strategy.
Collapse
Affiliation(s)
- E A F Bordini
- Department of Physiology and Pathology, Univ. Estadual Paulista - UNESP, Araraquara School of Dentistry, Humaitá Street, 1680, Araraquara, SP 14801-903, Brazil; Department of Operative Dentistry, Endodontics and Dental Materials, Sao Paulo University - USP, Bauru School of Dentistry, Al. Dr. Octávio Pinheiro Brizola, 9-75, Bauru, SP 17012-901, Brazil
| | - F B Cassiano
- Department of Operative Dentistry, Endodontics and Dental Materials, Sao Paulo University - USP, Bauru School of Dentistry, Al. Dr. Octávio Pinheiro Brizola, 9-75, Bauru, SP 17012-901, Brazil
| | - E S Bronze-Uhle
- Department of Operative Dentistry, Endodontics and Dental Materials, Sao Paulo University - USP, Bauru School of Dentistry, Al. Dr. Octávio Pinheiro Brizola, 9-75, Bauru, SP 17012-901, Brazil
| | - L Alamo
- Department of Operative Dentistry, Endodontics and Dental Materials, Sao Paulo University - USP, Bauru School of Dentistry, Al. Dr. Octávio Pinheiro Brizola, 9-75, Bauru, SP 17012-901, Brazil
| | - J Hebling
- Department of Orthodontics and Pediatric Dentistry, Univ. Estadual Paulista - UNESP, Araraquara School of Dentistry, Humaitá Street, 1680, Araraquara, SP 14801-903, Brazil
| | - C A de Souza Costa
- Department of Physiology and Pathology, Univ. Estadual Paulista - UNESP, Araraquara School of Dentistry, Humaitá Street, 1680, Araraquara, SP 14801-903, Brazil
| | - D G Soares
- Department of Operative Dentistry, Endodontics and Dental Materials, Sao Paulo University - USP, Bauru School of Dentistry, Al. Dr. Octávio Pinheiro Brizola, 9-75, Bauru, SP 17012-901, Brazil.
| |
Collapse
|
129
|
Qiu J, Fan X, Ding H, Zhao M, Xu T, Lei J, Ji B, Zhuang Z, Gao Q. Antenatal dexamethasone retarded fetal long bones growth and development by down-regulating of insulin-like growth factor 1 signaling in fetal rats. Hum Exp Toxicol 2022; 41:9603271211072870. [PMID: 35148621 DOI: 10.1177/09603271211072870] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Dexamethasone (DEX), a synthetic glucocorticoid, has been widely used as a medication for premature delivery. However, the side effects of antenatal DEX treatment on fetal bone development, as well as the underlying mechanisms still remain to be elucidated. Here, we aimed to explore the effects and the related mechanisms of antenatal DEX exposure during late pregnancy on fetal bone growth and development. METHODS Pregnant Sprague-Dawley rats were randomly divided into DEX group and vehicle group from gestational day 14 (GD14). Pregnant rats in DEX group were intraperitoneally injected once with DEX (200 µg/kg body weight) on GD14, 16, 18, and 20. The vehicle group rats were administered the same amount of normal saline at the same time. Pregnant rats were anesthetized at GD21 to harvest fetal femurs for analysis. RESULTS Antenatal DEX treatment delayed fetal skeletal growth via inhibiting extracellular matrix (ECM) synthesis and downregulating insulin-like growth factor 1 (IGF1) signaling. Several components of IGF1 signaling pathway, including IGF1 receptor, insulin receptor substrate, as well as serine-threonine protein kinase, were down-regulated in fetal growth plate chondrocytes following DEX treatment. CONCLUSION This study indicated that antenatal DEX treatment-retarded fetal skeletal growth was associated with the down-regulation of IGF1 signaling in growth plate chondrocytes, providing important information about the impact of antenatal DEX application four courses on premature infant.
Collapse
Affiliation(s)
- Junlan Qiu
- Institute for Fetology, 74566First Hospital of Soochow University, Suzhou, China.,Department of Oncology, 105860Second Affiliated Hospital of Soochow University, Suzhou, China.,Department of Oncology and Hematology, Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Xiaorong Fan
- Institute for Fetology, 74566First Hospital of Soochow University, Suzhou, China.,Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongmei Ding
- Institute for Fetology, 74566First Hospital of Soochow University, Suzhou, China
| | - Meng Zhao
- Institute for Fetology, 74566First Hospital of Soochow University, Suzhou, China
| | - Ting Xu
- Institute for Fetology, 74566First Hospital of Soochow University, Suzhou, China
| | - Jiahui Lei
- Institute for Fetology, 74566First Hospital of Soochow University, Suzhou, China
| | - Bingyu Ji
- Institute for Fetology, 74566First Hospital of Soochow University, Suzhou, China
| | - Zhixiang Zhuang
- Department of Oncology, 105860Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qinqin Gao
- Institute for Fetology, 74566First Hospital of Soochow University, Suzhou, China
| |
Collapse
|
130
|
van Santen VJB, Bastidas Coral AP, Hogervorst JMA, Klein-Nulend J, Bakker AD. Biologically Relevant In Vitro 3D-Model to Study Bone Regeneration Potential of Human Adipose Stem Cells. Biomolecules 2022; 12:biom12020169. [PMID: 35204670 PMCID: PMC8961519 DOI: 10.3390/biom12020169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 11/16/2022] Open
Abstract
Standard cell cultures may not predict the proliferation and differentiation potential of human mesenchymal stromal cells (MSCs) after seeding on a scaffold and implanting this construct in a bone defect. We aimed to develop a more biologically relevant in vitro 3D-model for preclinical studies on the bone regeneration potential of MSCs. Human adipose tissue-derived mesenchymal stromal cells (hASCs; five donors) were seeded on biphasic calcium phosphate (BCP) granules and cultured under hypoxia (1% O2) for 14 days with pro-inflammatory TNFα, IL4, IL6, and IL17F (10 mg/mL each) added during the first three days, simulating the early stages of repair (bone construct model). Alternatively, hASCs were cultured on plastic, under 20% O2 and without cytokines for 14 days (standard cell culture). After two days, the bone construct model decreased total DNA (3.9-fold), COL1 (9.8-fold), and RUNX2 expression (19.6-fold) and metabolic activity (4.6-fold), but increased VEGF165 expression (38.6-fold) in hASCs compared to standard cultures. After seven days, the bone construct model decreased RUNX2 expression (64-fold) and metabolic activity (2.3-fold), but increased VEGF165 (54.5-fold) and KI67 expression (5.7-fold) in hASCs compared to standard cultures. The effect of the bone construct model on hASC proliferation and metabolic activity could be largely mimicked by culturing on BCP alone (20% O2, no cytokines). The effect of the bone construct model on VEGF165 expression could be mimicked by culturing hASCs under hypoxia alone (plastic, no cytokines). In conclusion, we developed a new, biologically relevant in vitro 3D-model to study the bone regeneration potential of MSCs. Our model is likely more suitable for the screening of novel factors to enhance bone regeneration than standard cell cultures.
Collapse
|
131
|
Li T, Hou X, Huang Y, Wang C, Chen H, Yan C. In vitro and in silico anti-osteoporosis activities and underlying mechanisms of a fructan, ABW90-1, from Achyranthes bidentate. Carbohydr Polym 2022; 276:118730. [PMID: 34823766 DOI: 10.1016/j.carbpol.2021.118730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/25/2021] [Accepted: 09/30/2021] [Indexed: 11/18/2022]
Abstract
Achyranthes bidentata is a traditional Chinese medicine used to treat osteoporosis. AB90, a crude saccharide from A. bidentata, showed excellent osteoprotective effects in ovariectomized rats, and ABW90-1, an oligosaccharide purified from AB90, stimulated significant differentiation of osteoblasts. However, the osteogenic effects and underlying mechanisms of ABW90-1 have remained unknown. In the present study, we found that ABW90-1 significantly promoted ALP activity, mineralization, and the expression of osteogenic markers in MC3T3-E1 cells. ABW90-1 showed strong binding with the WNT signaling complex and BMP2 based on number of interactions, hydrogen bond length, and binding energy in silico. ABW90-1 significantly increased the expression of active-β-catenin, p-GSK-3β, LEF-1, BMP2, and p-SMAD1. Importantly, the osteogenic effects of ABW90-1 were partially suppressed by DKK-1 and Noggin, which are specific inhibitors of the WNT and BMP signaling pathways, respectively. Collectively, these findings suggest that ABW90-1 has osteogenic effects through crosstalk between WNT/β-catenin and BMP2/SMAD1 signaling pathways.
Collapse
Affiliation(s)
- Tianyu Li
- Clinical Pharmacy of the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510060, China; School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xin Hou
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yihua Huang
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Changsheng Wang
- Clinical Pharmacy of the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510060, China
| | - Haiyun Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chunyan Yan
- Clinical Pharmacy of the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510060, China; School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
132
|
Arora S, Cooper PR, Ratnayake JT, Friedlander LT, Rizwan SB, Seo B, Hussaini HM. A critical review of in vitro research methodologies used to study mineralization in human dental pulp cell cultures. Int Endod J 2022; 55 Suppl 1:3-13. [PMID: 35030284 PMCID: PMC9303903 DOI: 10.1111/iej.13684] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/11/2022] [Indexed: 11/27/2022]
Abstract
Background The pulp contains a resident population of stem cells which can be stimulated to differentiate in order to repair the tooth by generating a mineralized extracellular matrix. Over recent decades there has been considerable interest in utilizing in vitro cell culture models to study dentinogenesis, with the aim of developing regenerative endodontic procedures, particularly where some vital pulp tissue remains. Objectives The purpose of this review is to provide a structured oversight of in vitro research methodologies which have been used to study human pulp mineralization processes. Method The literature was screened in the PubMed database up to March 2021 to identify manuscripts reporting the use of human dental pulp cells to study mineralization. The dataset identified 343 publications initially which were further screened and consequently 166 studies were identified and it was methodologically mined for information on: i) study purpose, ii) source and characterization of cells, iii) mineralizing supplements and concentrations, and iv) assays and markers used to characterize mineralization and differentiation, and the data was used to write this narrative review. Results Most published studies aimed at characterizing new biological stimulants for mineralization as well as determining the effect of scaffolds and dental (bio)materials. In general, pulp cells were isolated by enzymatic digestion, although the pulp explant technique was also common. For enzymatic digestion, a range of enzymes and concentrations were utilized, although collagenase type I and dispase were the most frequent. Isolated cells were not routinely characterized using either fluorescence‐activated cell sorting (FACS) and magnetic‐activated cell sorting (MACS) approaches and there was little consistency in terming cultures as dental pulp cells or dental pulp stem cells. A combination of media supplements, at a range of concentrations, of dexamethasone, ascorbic acid and beta‐glycerophosphate, were frequently applied as the basis for the experimental conditions. Alizarin Red S (ARS) staining was the method of choice for assessment of mineralization at 21‐days. Alkaline phosphatase assay was relatively frequently applied, solely or in combination with ARS staining. Further assessment of differentiation status was performed using transcript or protein markers, with dentine sialophosphoprotein (DSPP), osteocalcin and dentine matrix protein‐1 (DMP ‐1), the most frequent. Discussion While this review highlights variability among experimental approaches, it does however identify a consensus experimental approach. Conclusion Standardization of experimental conditions and sustained research will significantly benefit endodontic patient outcomes in the future.
Collapse
Affiliation(s)
- Shelly Arora
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| | - Paul R Cooper
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| | - Jithendra T Ratnayake
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| | - Lara T Friedlander
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| | | | - Benedict Seo
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| | - Haizal M Hussaini
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin
| |
Collapse
|
133
|
Furlani F, Rossi A, Grimaudo MA, Bassi G, Giusto E, Molinari F, Lista F, Montesi M, Panseri S. Controlled Liposome Delivery from Chitosan-Based Thermosensitive Hydrogel for Regenerative Medicine. Int J Mol Sci 2022; 23:ijms23020894. [PMID: 35055097 PMCID: PMC8776110 DOI: 10.3390/ijms23020894] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/07/2023] Open
Abstract
This work describes the development of an injectable nanocomposite system based on a chitosan thermosensitive hydrogel combined with liposomes for regenerative medicine applications. Liposomes with good physicochemical properties are prepared and embedded within the chitosan network. The resulting nanocomposite hydrogel is able to provide a controlled release of the content from liposomes, which are able to interact with cells and be internalized. The cellular uptake is enhanced by the presence of a chitosan coating, and cells incubated with liposomes embedded within thermosensitive hydrogels displayed a higher cell uptake compared to cells incubated with liposomes alone. Furthermore, the gelation temperature of the system resulted to be equal to 32.6 °C; thus, the system can be easily injected in the target site to form a hydrogel at physiological temperature. Given the peculiar performance of the selected systems, the resulting thermosensitive hydrogels are a versatile platform and display potential applications as controlled delivery systems of liposomes for tissue regeneration.
Collapse
Affiliation(s)
- Franco Furlani
- National Research Council of Italy-Institute of Science and Technology for Ceramics (ISTEC-CNR), Via Granarolo 64, I-48018 Faenza, Italy; (A.R.); (M.A.G.); (G.B.); (E.G.); (M.M.)
- Correspondence: (F.F.); (S.P.); Tel.: +39-0546-699-776 (F.F.); +39-0546-699-785 (S.P.)
| | - Arianna Rossi
- National Research Council of Italy-Institute of Science and Technology for Ceramics (ISTEC-CNR), Via Granarolo 64, I-48018 Faenza, Italy; (A.R.); (M.A.G.); (G.B.); (E.G.); (M.M.)
| | - Maria Aurora Grimaudo
- National Research Council of Italy-Institute of Science and Technology for Ceramics (ISTEC-CNR), Via Granarolo 64, I-48018 Faenza, Italy; (A.R.); (M.A.G.); (G.B.); (E.G.); (M.M.)
| | - Giada Bassi
- National Research Council of Italy-Institute of Science and Technology for Ceramics (ISTEC-CNR), Via Granarolo 64, I-48018 Faenza, Italy; (A.R.); (M.A.G.); (G.B.); (E.G.); (M.M.)
| | - Elena Giusto
- National Research Council of Italy-Institute of Science and Technology for Ceramics (ISTEC-CNR), Via Granarolo 64, I-48018 Faenza, Italy; (A.R.); (M.A.G.); (G.B.); (E.G.); (M.M.)
| | - Filippo Molinari
- Army Medical Center, Scientific Department, I-00184 Rome, Italy; (F.M.); (F.L.)
| | - Florigio Lista
- Army Medical Center, Scientific Department, I-00184 Rome, Italy; (F.M.); (F.L.)
| | - Monica Montesi
- National Research Council of Italy-Institute of Science and Technology for Ceramics (ISTEC-CNR), Via Granarolo 64, I-48018 Faenza, Italy; (A.R.); (M.A.G.); (G.B.); (E.G.); (M.M.)
| | - Silvia Panseri
- National Research Council of Italy-Institute of Science and Technology for Ceramics (ISTEC-CNR), Via Granarolo 64, I-48018 Faenza, Italy; (A.R.); (M.A.G.); (G.B.); (E.G.); (M.M.)
- Correspondence: (F.F.); (S.P.); Tel.: +39-0546-699-776 (F.F.); +39-0546-699-785 (S.P.)
| |
Collapse
|
134
|
Chang H, Jiang T, Kou L, Li D, Yu X, Li Y, Zhang L. MiR-148a-3p Regulates Stem Cell Osteogenic Differentiation and Enamel Development by Targeting Runt-Related Transcription Factor 2 and E-cadherin <i>via</i> the Wnt1/β-catenin Signaling Pathway. J HARD TISSUE BIOL 2022. [DOI: 10.2485/jhtb.31.141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Huaiguang Chang
- Department of Stomatology, Ningbo College of Health Sciences
| | - Tingting Jiang
- Department of Prosthodontics, Yinzhou Stomatology Hospital
| | - Liang Kou
- Department of Stomatology, Ningbo College of Health Sciences
| | - Duo Li
- Department of Prosthodontics, Yinzhou Stomatology Hospital
| | - Xinchen Yu
- Department of Stomatology, Ningbo College of Health Sciences
| | - Youqin Li
- Department of Stomatology, Ningbo College of Health Sciences
| | - Lei Zhang
- Department of Stomatology, Ningbo College of Health Sciences
| |
Collapse
|
135
|
Sabino RM, Rau JV, De Bonis A, De Stefanis A, Curcio M, Teghil R, Popat KC. Manganese-containing Bioactive Glass Enhances Osteogenic Activity of TiO 2 Nanotube Arrays. APPLIED SURFACE SCIENCE 2021; 570:151163. [PMID: 34594060 PMCID: PMC8478254 DOI: 10.1016/j.apsusc.2021.151163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Titanium and its alloys are the most used biomaterials for orthopedic and dental applications. However, up to 10% of these medical devices still fail, mostly due to implant loosening and suboptimal integration at the implant site. The biomaterial surface plays a critical role in promoting osseointegration, which can reduce the risk of device failure. In this study, we propose a novel surface modification on titanium to improve osteogenic differentiation by depositing manganese-containing bioactive glass (BG) on TiO2 nanotube arrays. The surfaces were characterized by scanning electron microscopy, energy dispersive X-ray spectrometer, contact angle goniometry, and X-ray photoelectron spectroscopy. Cell toxicity, viability, adhesion, and proliferation of adipose-derived stem cells on the surfaces were investigated up to 7 days. To evaluate the osteogenic properties of the surfaces, alkaline phosphatase activity, total protein, osteocalcin expression, and calcium deposition were quantified up to 28 days. The results indicate that TiO2 nanotube arrays modified with BG promote cell growth and induce increased osteocalcin and calcium contents when compared to unmodified TiO2 nanotube arrays. The deposition of manganese-containing bioactive glass onto TiO2 nanotubes demonstrates the ability to enhance osteogenic activity on titanium, showing great potential for use in orthopedic and dental implants.
Collapse
Affiliation(s)
- Roberta M. Sabino
- School of Advanced Materials Discovery, Colorado State University, Fort Collins, USA
| | - Julietta V. Rau
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (ISM-CNR), Via del Fosso del Cavaliere 100, 00133 Rome, Italy
- I.M. Sechenov First Moscow State Medical University, Institute of Pharmacy, Department of Analytical, Physical and Colloid Chemistry, Trubetskaya 8, build. 2, 119991 Moscow, Russia
| | - Angela De Bonis
- Dipartimento di Scienze, Università della Basilicata, Via dell’Ateneo Lucano, 10-85100 Potenza, Italy
| | - Adriana De Stefanis
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (ISM-CNR), Montelibretti Unit, Via Salaria km 29.300, 00015 Monterotondo Scalo, Italy
| | - Mariangela Curcio
- Dipartimento di Scienze, Università della Basilicata, Via dell’Ateneo Lucano, 10-85100 Potenza, Italy
| | - Roberto Teghil
- Dipartimento di Scienze, Università della Basilicata, Via dell’Ateneo Lucano, 10-85100 Potenza, Italy
| | - Ketul C. Popat
- School of Advanced Materials Discovery, Colorado State University, Fort Collins, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, USA
- Department of Mechanical Engineering, Colorado State University, Fort Collins, USA
| |
Collapse
|
136
|
Rashid U, Yousaf A, Yaqoob M, Saba E, Moaeen-Ud-Din M, Waseem S, Becker SK, Sponder G, Aschenbach JR, Sandhu MA. Characterization and differentiation potential of mesenchymal stem cells isolated from multiple canine adipose tissue sources. BMC Vet Res 2021; 17:388. [PMID: 34922529 PMCID: PMC8684202 DOI: 10.1186/s12917-021-03100-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) are undifferentiated cells that can give rise to a mesoderm lineage. Adipose-derived MSCs are an easy and accessible source for MSCs isolation, although each source of MSC has its own advantages and disadvantages. Our study identifies a promising source for the isolation and differentiation of canines MSCs. For this purpose, adipose tissue from inguinal subcutaneous (SC), perirenal (PR), omental (OM), and infrapatellar fat pad (IPFP) was isolated and processed for MSCs isolation. In the third passage, MSCs proliferation/metabolism, surface markers expression, in vitro differentiation potential and quantitative reverse transcription PCR (CD73, CD90, CD105, PPARγ, FabP4, FAS, SP7, Osteopontin, and Osteocalcin) were evaluated. Results Our results showed that MSCs derived from IPFP have a higher proliferation rate, while OM-derived MSCs have higher cell metabolism. In addition, MSCs from all adipose tissue sources showed positive expression of CD73 (NT5E), CD90 (THY1), CD105 (ENDOGLIN), and very low expression of CD45. The isolated canine MSCs were successfully differentiated into adipogenic and osteogenic lineages. The oil-red-O quantification and adipogenic gene expression (FAS, FabP4, and PPARγ) were higher in OM-derived cells, followed by IPFP-MSCs. Similarly, in osteogenic differentiation, alkaline phosphatase activity and osteogenic gene (SP7 and Osteocalcin) expression were higher in OM-derived MSCs, while osteopontin expression was higher in PR-derived MSCs. Conclusion In summary, among all four adipose tissue sources, OM-derived MSCs have better differentiation potential toward adipo- and osteogenic lineages, followed by IPFP-MSCs. Interestingly, among all adipose tissue sources, MSCs derived from IPFP have the maximum proliferation potential. The characterization and differentiation potential of canine MSCs isolated from four different adipose tissue sources are useful to assess their potential for application in regenerative medicine.
Collapse
Affiliation(s)
- Usman Rashid
- Department of Clinical Studies, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi, 46300, Pakistan
| | - Arfan Yousaf
- Department of Clinical Studies, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi, 46300, Pakistan
| | - Muhammad Yaqoob
- Department of Clinical Studies, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi, 46300, Pakistan
| | - Evelyn Saba
- Department of Veterinary Biomedical Sciences, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi, 46300, Pakistan
| | - Muhammad Moaeen-Ud-Din
- Department of Animal Breeding and Genetics, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi, 46300, Pakistan
| | | | - Sandra K Becker
- Institute of Veterinary-Physiology, Freie Universität Berlin, Berlin, Germany
| | - Gerhard Sponder
- Institute of Veterinary-Physiology, Freie Universität Berlin, Berlin, Germany
| | - Jörg R Aschenbach
- Institute of Veterinary-Physiology, Freie Universität Berlin, Berlin, Germany
| | - Mansur Abdullah Sandhu
- Department of Veterinary Biomedical Sciences, Faculty of Veterinary and Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi, 46300, Pakistan.
| |
Collapse
|
137
|
Brito Barrera YA, Husteden C, Alherz J, Fuhrmann B, Wölk C, Groth T. Extracellular matrix-inspired surface coatings functionalized with dexamethasone-loaded liposomes to induce osteo- and chondrogenic differentiation of multipotent stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112516. [PMID: 34857295 DOI: 10.1016/j.msec.2021.112516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 10/20/2022]
Abstract
Biomimetic surface coatings can be combined with conventional implants to mimic the extracellular matrix (ECM) of the surrounding tissue to make them more biocompatible. Layer-by-layer technique (LbL) can be used for making surface coatings by alternating adsorption of polyanions and polycations from aqueous solutions without need of chemical reactions. Here, polyelectrolyte multilayer (PEM) systems is made of hyaluronic acid (HA) as polyanion and Collagen I (Col) as polycation to mimic the ECM of connective tissue. The PEM are combined with dexamethasone (Dex)-loaded liposomes to achieve a local delivery and protection of this drug for stimulation of osteo- and chondrogenic differentiation of multipotent stem cells. The liposomes possess a positive surface charge that is required for immobilization on the PEM. The surface properties of PEM system show a positive zeta potential after liposome adsorption and a decrease in wettability, both promoting cell adhesion and spreading of C3H10T1/2 multipotent embryonic mouse fibroblasts. Differentiation of C3H10T1/2 was more prominent on the PEM system with embedded Dex-loaded liposomes compared to the basal PEM system and the use of free Dex-loaded liposomes in the supernatant. This was evident by immunohistochemical staining and an upregulation of the expression of genes, which play a key role in osteogenesis (RunX2, ALP, Osteocalcin (OCN)) and chondrogenesis (Sox9, aggrecan (ACAN), collagen type II), determined by quantitative Real-time polymerase chain reaction (qRT-PCR) after 21 days. These findings indicate that the designed liposome-loaded PEM system have high potential for use as drug delivery systems for implant coatings that can induce bone and cartilage differentiation needed for example in osteochondral implants.
Collapse
Affiliation(s)
- Yazmin A Brito Barrera
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Heinrich-Damerow-Strasse 4, 06120 Halle (Saale), Germany
| | - Catharina Husteden
- Medicinal Chemistry Department, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Jumanah Alherz
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Heinrich-Damerow-Strasse 4, 06120 Halle (Saale), Germany
| | - Bodo Fuhrmann
- Interdisciplinary Center of Materials Science, Martin Luther University Halle-Wittenberg, D-06099 Halle (Saale), Germany
| | - Christian Wölk
- Pharmaceutical Technology, Institute of Pharmacy, Faculty of Medicine, Leipzig University, 04317 Leipzig, Germany
| | - Thomas Groth
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Heinrich-Damerow-Strasse 4, 06120 Halle (Saale), Germany; Interdisciplinary Center of Materials Science, Martin Luther University Halle-Wittenberg, D-06099 Halle (Saale), Germany; Laboratory of Biomedical Nanotechnologies, Institute of Bionic Technologies and Engineering, I.M. Sechenov First Moscow State University, 119991, Trubetskaya street 8, Moscow, Russian Federation.
| |
Collapse
|
138
|
Wei W, Dai H. Articular cartilage and osteochondral tissue engineering techniques: Recent advances and challenges. Bioact Mater 2021; 6:4830-4855. [PMID: 34136726 PMCID: PMC8175243 DOI: 10.1016/j.bioactmat.2021.05.011] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/20/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
In spite of the considerable achievements in the field of regenerative medicine in the past several decades, osteochondral defect regeneration remains a challenging issue among diseases in the musculoskeletal system because of the spatial complexity of osteochondral units in composition, structure and functions. In order to repair the hierarchical tissue involving different layers of articular cartilage, cartilage-bone interface and subchondral bone, traditional clinical treatments including palliative and reparative methods have showed certain improvement in pain relief and defect filling. It is the development of tissue engineering that has provided more promising results in regenerating neo-tissues with comparable compositional, structural and functional characteristics to the native osteochondral tissues. Here in this review, some basic knowledge of the osteochondral units including the anatomical structure and composition, the defect classification and clinical treatments will be first introduced. Then we will highlight the recent progress in osteochondral tissue engineering from perspectives of scaffold design, cell encapsulation and signaling factor incorporation including bioreactor application. Clinical products for osteochondral defect repair will be analyzed and summarized later. Moreover, we will discuss the current obstacles and future directions to regenerate the damaged osteochondral tissues.
Collapse
Affiliation(s)
- Wenying Wei
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
- International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, 430070, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
- Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu Hydrogen Valley, Foshan, 528200, China
| |
Collapse
|
139
|
Guo P, Liu X, Zhang P, He Z, Li Z, Alini M, Richards RG, Grad S, Stoddart MJ, Zhou G, Zou X, Chan D, Tian W, Chen D, Gao M, Zhou Z, Liu S. A single-cell transcriptome of mesenchymal stromal cells to fabricate bioactive hydroxyapatite materials for bone regeneration. Bioact Mater 2021; 9:281-298. [PMID: 34820571 PMCID: PMC8586438 DOI: 10.1016/j.bioactmat.2021.08.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/01/2021] [Accepted: 08/05/2021] [Indexed: 01/07/2023] Open
Abstract
The osteogenic microenvironment of bone-repairing materials plays a key role in accelerating bone regeneration but remains incompletely defined, which significantly limits the application of such bioactive materials. Here, the transcriptional landscapes of different osteogenic microenvironments, including three-dimensional (3D) hydroxyapatite (HA) scaffolds and osteogenic medium (OM), for mesenchymal stromal cells (MSCs) in vitro were mapped at single-cell resolution. Our findings suggested that an osteogenic process reminiscent of endochondral ossification occurred in HA scaffolds through sequential activation of osteogenic-related signaling pathways, along with inflammation and angiogenesis, but inhibition of adipogenesis and fibrosis. Moreover, we revealed the mechanism during OM-mediated osteogenesis involves the ZBTB16 and WNT signaling pathways. Heterogeneity of MSCs was also demonstrated. In vitro ossification of LRRC75A+ MSCs was shown to have better utilization of WNT-related ossification process, and PCDH10+ MSCs with superiority in hydroxyapatite-related osteogenic process. These findings provided further understanding of the cellular activity modulated by OM conditions and HA scaffolds, providing new insights for the improvement of osteogenic biomaterials. This atlas provides a blueprint for research on MSC heterogeneity and the osteogenic microenvironment of HA scaffolds and a database reference for the application of bioactive materials for bone regeneration.
Collapse
Affiliation(s)
- Peng Guo
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xizhe Liu
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Penghui Zhang
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhongyuan He
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen Li
- AO Research Institute Davos, Davos, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | | | | | | - Guangqian Zhou
- Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Health Sciences Center, Shenzhen University, Shenzhen, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Danny Chan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Wei Tian
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopaedics and Traumatology, Beijing JiShuiTan Hospital, Beijing, China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopaedics and Traumatology, Beijing JiShuiTan Hospital, Beijing, China
- Corresponding author.
| | - Manman Gao
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Sport Medicine, Inst Translat Med, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Corresponding author. Department of Sport Medicine, Inst Translat Med, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China.
| | - Zhiyu Zhou
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Corresponding author. Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Shaoyu Liu
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
140
|
Font Tellado S, Delgado JA, Poh SPP, Zhang W, García-Vallés M, Martínez S, Gorustovich A, Morejón L, van Griensven M, Balmayor ER. Phosphorous pentoxide-free bioactive glass exhibits dose-dependent angiogenic and osteogenic capacities which are retained in glass polymeric composite scaffolds. Biomater Sci 2021; 9:7876-7894. [PMID: 34676835 DOI: 10.1039/d1bm01311d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bioactive glasses (BGs) are attractive materials for bone tissue engineering because of their bioactivity and osteoinductivity. In this study, we report the synthesis of a novel phosphorous pentoxide-free, silicate-based bioactive glass (52S-BG) composed of 52.1% SiO2, 23.2% Na2O and 22.6% CaO (wt%). The glass was thoroughly characterized. The biocompatibility and osteogenic properties of 52S-BG particles were analyzed in vitro with human adipose-derived mesenchymal stem cells (AdMSCs) and human osteoblasts. 52S-BG particles were biocompatible and induced mineralized matrix deposition and the expression of osteogenic markers (RunX2, alkaline phosphatase, osteocalcin, osteopontin, collagen I) and the angiogenic marker vascular endothelial growth factor (VEGF). Angiogenic properties were additionally confirmed in a zebrafish embryo model. 52S-BG was added to poly-ε-caprolactone (PCL) to obtain a composite with 10 wt% glass content. Composite PCL/52S-BG scaffolds were fabricated by additive manufacturing and displayed high porosity (76%) and pore interconnectivity. The incorporation of 52S-BG particles increased the Young's modulus of PCL scaffolds from 180 to 230 MPa. AdMSC seeding efficiency and proliferation were higher in PCL/52S-BG compared to PCL scaffolds, indicating improved biocompatibility. Finally, 52S-BG incorporation improved the scaffolds' osteogenic and angiogenic properties by increasing mineral deposition and inducing relevant gene expression and VEGF protein secretion. Overall, 52S-BG particles and PCL/52S-BG composites may be attractive for diverse bone engineering applications requiring concomitant angiogenic properties.
Collapse
Affiliation(s)
- Sonia Font Tellado
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - José Angel Delgado
- Center for Biomaterials, University of Havana, 10400 Havana, Cuba.,Universitat Internacional de Catalunya, 08195 Barcelona, Spain
| | - Su Ping Patrina Poh
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, 13353 Berlin, Germany
| | - Wen Zhang
- Institute of Molecular Immunology and Experimental Oncology, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany.,Ethris GmbH, 82152 Planegg, Germany
| | - Maite García-Vallés
- Mineralogy, Petrology and Applied Geology Department, University of Barcelona, 08028 Barcelona, Spain
| | - Salvador Martínez
- Mineralogy, Petrology and Applied Geology Department, University of Barcelona, 08028 Barcelona, Spain
| | - Alejandro Gorustovich
- Interdisciplinary Materials Group-IESIING-UCASAL, INTECIN UBA-CONICET, A4400EDD Salta, Argentina
| | - Lizette Morejón
- Center for Biomaterials, University of Havana, 10400 Havana, Cuba
| | - Martijn van Griensven
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany.,cBITE, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6200 MD Maastricht, the Netherlands
| | - Elizabeth Rosado Balmayor
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany.,IBE, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6200 MD Maastricht, the Netherlands.
| |
Collapse
|
141
|
Remmers SJA, de Wildt BWM, Vis MAM, Spaander ESR, de Vries RBM, Ito K, Hofmann S. Osteoblast-osteoclast co-cultures: A systematic review and map of available literature. PLoS One 2021; 16:e0257724. [PMID: 34735456 PMCID: PMC8568160 DOI: 10.1371/journal.pone.0257724] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/21/2021] [Indexed: 01/22/2023] Open
Abstract
Drug research with animal models is expensive, time-consuming and translation to clinical trials is often poor, resulting in a desire to replace, reduce, and refine the use of animal models. One approach to replace and reduce the use of animal models is to use in vitro cell-culture models. To study bone physiology, bone diseases and drugs, many studies have been published using osteoblast-osteoclast co-cultures. The use of osteoblast-osteoclast co-cultures is usually not clearly mentioned in the title and abstract, making it difficult to identify these studies without a systematic search and thorough review. As a result, researchers are all developing their own methods, leading to conceptually similar studies with many methodological differences and, as a consequence, incomparable results. The aim of this study was to systematically review existing osteoblast-osteoclast co-culture studies published up to 6 January 2020, and to give an overview of their methods, predetermined outcome measures (formation and resorption, and ALP and TRAP quantification as surrogate markers for formation and resorption, respectively), and other useful parameters for analysis. Information regarding these outcome measures was extracted and collected in a database, and each study was further evaluated on whether both the osteoblasts and osteoclasts were analyzed using relevant outcome measures. From these studies, additional details on methods, cells and culture conditions were extracted into a second database to allow searching on more characteristics. The two databases presented in this publication provide an unprecedented amount of information on cells, culture conditions and analytical techniques for using and studying osteoblast-osteoclast co-cultures. They allow researchers to identify publications relevant to their specific needs and allow easy validation and comparison with existing literature. Finally, we provide the information and tools necessary for others to use, manipulate and expand the databases for their needs.
Collapse
Affiliation(s)
- Stefan J. A. Remmers
- Department of Biomedical Engineering and the Institute of Complex Molecular Systems, Orthopaedic Biomechanics, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Bregje W. M. de Wildt
- Department of Biomedical Engineering and the Institute of Complex Molecular Systems, Orthopaedic Biomechanics, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Michelle A. M. Vis
- Department of Biomedical Engineering and the Institute of Complex Molecular Systems, Orthopaedic Biomechanics, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Eva S. R. Spaander
- Department of Biomedical Engineering and the Institute of Complex Molecular Systems, Orthopaedic Biomechanics, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Rob B. M. de Vries
- Department for Health Evidence, SYRCLE, Radboud Institute for Health Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Keita Ito
- Department of Biomedical Engineering and the Institute of Complex Molecular Systems, Orthopaedic Biomechanics, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Sandra Hofmann
- Department of Biomedical Engineering and the Institute of Complex Molecular Systems, Orthopaedic Biomechanics, Eindhoven University of Technology, Eindhoven, The Netherlands
- * E-mail:
| |
Collapse
|
142
|
Xiao T, Fan L, Liu R, Huang X, Wang S, Xiao L, Pang Y, Li D, Liu J, Min Y. Fabrication of Dexamethasone-Loaded Dual-Metal-Organic Frameworks on Polyetheretherketone Implants with Bacteriostasis and Angiogenesis Properties for Promoting Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2021; 13:50836-50850. [PMID: 34689546 DOI: 10.1021/acsami.1c18088] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Polyetheretherketone (PEEK) is a biocompatible polymer, but its clinical application is largely limited due to its inert surface. To solve this problem, a multifunctional PEEK implant is urgently fabricated. In this work, a dual-metal-organic framework (Zn-Mg-MOF74) coating is bonded to PEEK using a mussel-inspired polydopamine interlayer to prepare the coating, and then, dexamethasone (DEX) is loaded on the coating surface. The PEEK surface with the multifunctional coating provides superior hydrophilicity and favorable stability and forms an alkaline microenvironment when Mg2+, Zn2+, 2,5-dihydroxyterephthalic acid, and DEX are released due to the coating degradation. In vitro results showed that the multifunctional coating has strong antibacterial ability against both Escherichia coli and Staphylococcus aureus; it also improves human umbilical vein endothelial cell angiogenic ability and enhances rat bone marrow mesenchymal stem cell osteogenic differentiation activity. Furthermore, the in vivo rat subcutaneous infection model, chicken chorioallantoic membrane model, and rat femoral drilling model verify that the PEEK implant coated with the multifunctional coating has strong antibacterial and angiogenic ability and promotes the formation of new bone around the implant with a stronger bone-implant interface. Our findings indicate that DEX loaded on the Zn-Mg-MOF74 coating-modified PEEK implant with bacteriostasis, angiogenesis, and osteogenesis properties has great clinical application potential as bone graft materials.
Collapse
Affiliation(s)
- Tianhua Xiao
- School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China
| | - Lei Fan
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Rongtao Liu
- School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China
| | - Xingwen Huang
- School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China
| | - Shihuan Wang
- Child Developmental & Behavioral Center, Third Affiliated Hospital of Sun Yat-sen University, No.600, Tianhe Road, Guangzhou 510630, China
| | - Liangang Xiao
- School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China
| | - Yiyu Pang
- School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China
| | - Da Li
- School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China
| | - Jia Liu
- School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China
| | - Yonggang Min
- School of Materials and Energy, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
143
|
Wojcik M, Kazimierczak P, Vivcharenko V, Koziol M, Przekora A. Effect of Vitamin C/Hydrocortisone Immobilization within Curdlan-Based Wound Dressings on In Vitro Cellular Response in Context of the Management of Chronic and Burn Wounds. Int J Mol Sci 2021; 22:ijms222111474. [PMID: 34768905 PMCID: PMC8583867 DOI: 10.3390/ijms222111474] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/13/2021] [Accepted: 10/22/2021] [Indexed: 01/08/2023] Open
Abstract
Bioactive dressings are usually produced using natural or synthetic polymers. Recently, special attention has been paid to β-glucans that act as immunomodulators and have pro-healing properties. The aim of this research was to use β-1,3-glucan (curdlan) as a base for the production of bioactive dressing materials (curdlan/agarose and curdlan/chitosan) that were additionally enriched with vitamin C and/or hydrocortisone to improve healing of chronic and burn wounds. The secondary goal of the study was to compressively evaluate biological properties of the biomaterials. In this work, it was shown that vitamin C/hydrocortisone-enriched biomaterials exhibited faster vitamin C release profile than hydrocortisone. Consecutive release of the drugs is a desired phenomenon since it protects wounds against accumulation of high and toxic concentrations of the bioactive molecules. Moreover, biomaterials showed gradual release of low doses of the hydrocortisone, which is beneficial during management of burn wounds with hypergranulation tissue. Among all tested variants of biomaterials, dressing materials enriched with hydrocortisone and a mixture of vitamin C/hydrocortisone showed the best therapeutic potential since they had the ability to significantly reduce MMP-2 synthesis by macrophages and increase TGF-β1 release by skin cells. Moreover, materials containing hydrocortisone and its blend with vitamin C stimulated type I collagen deposition by fibroblasts and positively affected their migration and proliferation. Results of the experiments clearly showed that the developed biomaterials enriched with bioactive agents may be promising dressings for the management of non-healing chronic and burn wounds.
Collapse
Affiliation(s)
- Michal Wojcik
- Independent Unit of Tissue Engineering and Regenerative Medicine, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (M.W.); (P.K.); (V.V.)
| | - Paulina Kazimierczak
- Independent Unit of Tissue Engineering and Regenerative Medicine, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (M.W.); (P.K.); (V.V.)
| | - Vladyslav Vivcharenko
- Independent Unit of Tissue Engineering and Regenerative Medicine, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (M.W.); (P.K.); (V.V.)
| | - Malgorzata Koziol
- Department of Medical Microbiology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland;
| | - Agata Przekora
- Independent Unit of Tissue Engineering and Regenerative Medicine, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (M.W.); (P.K.); (V.V.)
- Correspondence: ; Tel.: +48-81-448-70-26
| |
Collapse
|
144
|
Lee S, Chae DS, Song BW, Lim S, Kim SW, Kim IK, Hwang KC. ADSC-Based Cell Therapies for Musculoskeletal Disorders: A Review of Recent Clinical Trials. Int J Mol Sci 2021; 22:ijms221910586. [PMID: 34638927 PMCID: PMC8508846 DOI: 10.3390/ijms221910586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 01/04/2023] Open
Abstract
Recently published clinical trials involving the use of adipose-derived stem cells (ADSCs) indicated that approximately one-third of the studies were conducted on musculoskeletal disorders (MSD). MSD refers to a wide range of degenerative conditions of joints, bones, and muscles, and these conditions are the most common causes of chronic disability worldwide, being a major burden to the society. Conventional treatment modalities for MSD are not sufficient to correct the underlying structural abnormalities. Hence, ADSC-based cell therapies are being tested as a form of alternative, yet more effective, therapies in the management of MSDs. Therefore, in this review, MSDs subjected to the ADSC-based therapy were further categorized as arthritis, craniomaxillofacial defects, tendon/ligament related disorders, and spine disorders, and their brief characterization as well as the corresponding conventional therapeutic approaches with possible mechanisms with which ADSCs produce regenerative effects in disease-specific microenvironments were discussed to provide an overview of under which circumstances and on what bases the ADSC-based cell therapy was implemented. Providing an overview of the current status of ADSC-based cell therapy on MSDs can help to develop better and optimized strategies of ADSC-based therapeutics for MSDs as well as help to find novel clinical applications of ADSCs in the near future.
Collapse
Affiliation(s)
- Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Dong-Sik Chae
- Department of Orthopedic Surgery, International St. Mary’s Hospital, Catholic Kwandong University, Gangneung 210-701, Korea;
| | - Byeong-Wook Song
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Il-Kwon Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
- Correspondence: (I.-K.K.); (K.-C.H.); Fax: +82-32-290-2774 (K.-C.H.)
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
- Correspondence: (I.-K.K.); (K.-C.H.); Fax: +82-32-290-2774 (K.-C.H.)
| |
Collapse
|
145
|
Phloroglucinol-enhanced whey protein isolate hydrogels with antimicrobial activity for tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112412. [PMID: 34579921 DOI: 10.1016/j.msec.2021.112412] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/12/2021] [Accepted: 08/30/2021] [Indexed: 12/28/2022]
Abstract
Aging populations in developed countries will increase the demand for implantable materials to support tissue regeneration. Whey Protein Isolate (WPI), derived from dairy industry by-products, can be processed into hydrogels with the following desirable properties for applications in tissue engineering: (i) ability to support adhesion and growth of cells; (ii) ease of sterilization by autoclaving and (iii) ease of incorporation of poorly water-soluble drugs with antimicrobial activity, such as phloroglucinol (PG), the fundamental phenolic subunit of marine polyphenols. In this study, WPI hydrogels were enriched with PG at concentrations between 0 and 20% w/v. PG solubilization in WPI hydrogels is far higher than in water. Enrichment with PG did not adversely affect mechanical properties, and endowed antimicrobial activity against a range of bacteria which occur in healthcare-associated infections (HAI). WPI-PG hydrogels supported the growth of, and collagen production by human dental pulp stem cells and - to a lesser extent - of osteosarcoma-derived MG-63 cells. In summary, enrichment of WPI with PG may be a promising strategy to prevent microbial contamination while still promoting stem cell attachment and growth.
Collapse
|
146
|
Amr M, Mallah A, Abusharkh H, Van Wie B, Gozen A, Mendenhall J, Idone V, Tingstad E, Abu-Lail NI. In vitro effects of nutraceutical treatment on human osteoarthritic chondrocytes of females of different age and weight groups. J Nutr Sci 2021; 10:e82. [PMID: 34616553 PMCID: PMC8477349 DOI: 10.1017/jns.2021.79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/18/2021] [Accepted: 08/31/2021] [Indexed: 11/05/2022] Open
Abstract
The in vitro effects of four nutraceuticals, catechin hydrate, gallic acid, α-tocopherol and ascorbic acid, on the ability of human osteoarthritic chondrocytes of two female obese groups to form articular cartilage (AC) tissues and to reduce inflammation were investigated. Group 1 represented thirteen females in the 50-69 years old range, an average weight of 100 kg and an average body mass index (BMI) of 34⋅06 kg/m2. Group 2 was constituted of three females in the 70-80 years old range, an average weight of 75 kg and an average BMI of 31⋅43 kg/m2. The efficacy of nutraceuticals was assessed in monolayer cultures using histological, colorimetric and mRNA gene expression analyses. AC engineered tissues of group 1 produced less total collagen and COL2A1 (38-fold), and higher COL10A1 (2⋅7-fold), MMP13 (50-fold) and NOS2 (15-fold) mRNA levels than those of group 2. In comparison, engineered tissues of group 1 had a significant decrease in NO levels from day 1 to day 21 (2⋅6-fold), as well as higher mRNA levels of FOXO1 (2-fold) and TNFAIP6 (16-fold) compared to group 2. Catechin hydrate decreased NO levels significantly in group 1 (1⋅5-fold) while increasing NO levels significantly in group 2 (3⋅8-fold). No differences from the negative control were observed in the presence of other nutraceuticals for either group. In conclusion, engineered tissues of the younger but heavier patients responded better to nutraceuticals than those from the older but leaner study participants. Finally, cells of group 2 formed better AC tissues with less inflammation and better extracellular matrix than cells of group 1.
Collapse
Key Words
- AA or vitamin C, ascorbic acid
- AC, articular cartilage
- Age
- Articualr cartilage
- BMI, body mass index
- C, catechin hydrate
- Catechin hydrate
- ECM, extracellular matrix
- G, gallic acid
- GAG, glycosaminoglycan
- MMP, metalloproteinase
- NO, nitric oxide
- NOS, NO Synthase
- Nutraceuticals
- OA, osteoarthritis
- Osteoarthritis
- TKR, total knee replacement
- TNF-α, tumour necrosis alpha
- Weight
- hAChs, human articular chondrocytes
- α or vitamin E, α-tocopherol
Collapse
Affiliation(s)
- Mahmoud Amr
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX78249, USA
| | - Alia Mallah
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX78249, USA
| | - Haneen Abusharkh
- Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA99164-6515, USA
| | - Bernard Van Wie
- Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA99164-6515, USA
| | - Arda Gozen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA99164-2920, USA
| | - Juana Mendenhall
- Department of Chemistry, Morehouse College, Atlanta, GA30314, USA
| | - Vincent Idone
- Regeneron Pharmaceuticals Inc, Tarrytown, NY10591, USA
| | - Edwin Tingstad
- Inland Orthopedic Surgery and Sports Clinic, Pullman, WA99163, USA
| | - Nehal I. Abu-Lail
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX78249, USA
| |
Collapse
|
147
|
Genetic profiling of human bone marrow and adipose tissue-derived mesenchymal stem cells reveals differences in osteogenic signaling mediated by graphene. J Nanobiotechnology 2021; 19:285. [PMID: 34551771 PMCID: PMC8459567 DOI: 10.1186/s12951-021-01024-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/02/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND In the last decade, graphene surfaces have consistently supported osteoblast development of stem cells, holding promise as a therapeutic implant for degenerative bone diseases. However, until now no study has specifically examined the genetic changes when stem cells undergo osteogenic differentiation on graphene. RESULTS In this study, we provide a detailed overview of gene expressions when human mesenchymal stem cells (MSCs) derived from either adipose tissue (AD-MSCs) or bone marrow (BM-MSCs), are cultured on graphene. Genetic expressions were measured using osteogenic RT2 profiler PCR arrays and compared either over time (7 or 21 days) or between each cell source at each time point. Genes were categorized as either transcriptional regulation, osteoblast-related, extracellular matrix, cellular adhesion, BMP and SMAD signaling, growth factors, or angiogenic factors. Results showed that both MSC sources cultured on low oxygen graphene surfaces achieved osteogenesis by 21 days and expressed specific osteoblast markers. However, each MSC source cultured on graphene did have genetically different responses. When compared between each other, we found that genes of BM-MSCs were robustly expressed, and more noticeable after 7 days of culturing, suggesting BM-MSCs initiate osteogenesis at an earlier time point than AD-MSCs on graphene. Additionally, we found upregulated angiogenic markers in both MSCs sources, suggesting graphene could simultaneously attract the ingrowth of blood vessels in vivo. Finally, we identified several novel targets, including distal-less homeobox 5 (DLX5) and phosphate-regulating endopeptidase homolog, X-linked (PHEX). CONCLUSIONS Overall, this study shows that graphene genetically supports differentiation of both AD-MSCs and BM-MSCs but may involve different signaling mechanisms to achieve osteogenesis. Data further demonstrates the lack of aberrant signaling due to cell-graphene interaction, strengthening the application of specific form and concentration of graphene nanoparticles in bone tissue engineering.
Collapse
|
148
|
Abd Rahman F. Gene expression profiling on effect of aspirin on osteogenic differentiation of periodontal ligament stem cells. BDJ Open 2021; 7:35. [PMID: 34531365 PMCID: PMC8446061 DOI: 10.1038/s41405-021-00090-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 01/07/2023] Open
Abstract
Periodontal ligament (PDL) contains a unique population of mesenchymal stem cells (MSCs), also known as PDL stem cells (PDLSCs). The regenerative properties of PDLSCs hold great potential for its use in stem cells based therapy, particularly for periodontal or bone regeneration. The present study investigated the global gene expression profile in PDLSCs during osteogenic differentiation. MSCs from PDL were isolated from normal permanent human teeth (n = 3). Microarray analysis was used to study the effects of ASA (200, 500, and 1000 μM) on the gene expression profiles in PDLSCs during osteogenic differentiation. Microarray study revealed that ASA was able to modulate PDLSCs gene expression profile. At 200 µM, 315 genes were dysregulated genes (DE), involving 151 upregulated and 164 downregulated genes. At 500 µM, 794 genes were DE, involving of 364 upregulated and 430 downregulated genes. At 1000 µM, the number of DE genes increased to 2035, of which 735 were upregulated and 1300 were downregulated. Bioinformatics analyses of the gene expression data revealed that the majority of DE genes (for 500 and 1000 µM ASA treatment) are involved in osteogenic differentiation. The gene network analysis was carried out using Ingenuity Pathway Analysis (IPA) software, and this revealed that the number of gene groups involved in cell adhesion and extracellular matrix components were increased. This study indicated that ASA could enhance PDLSCs functions and provide evidence for the potential use of ASA with PDLSCs for regenerative dentistry applications, particularly in the areas of periodontal health and regeneration. Periodontal ligament stem cells (PDLSCs) Aspirin (ASA) Microarray Osteogenic.
Collapse
Affiliation(s)
- Fazliny Abd Rahman
- Faculty of Dentistry, SEGi University, Kota Damansara, 47810, Petaling Jaya, Selangor, Malaysia.
| |
Collapse
|
149
|
Kang MS, Jang HJ, Lee SH, Lee JE, Jo HJ, Jeong SJ, Kim B, Han DW. Potential of Carbon-Based Nanocomposites for Dental Tissue Engineering and Regeneration. MATERIALS (BASEL, SWITZERLAND) 2021; 14:5104. [PMID: 34501203 PMCID: PMC8434078 DOI: 10.3390/ma14175104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022]
Abstract
While conventional dental implants focus on mechanical properties, recent advances in functional carbon nanomaterials (CNMs) accelerated the facilitation of functionalities including osteoinduction, osteoconduction, and osseointegration. The surface functionalization with CNMs in dental implants has emerged as a novel strategy for reinforcement and as a bioactive cue due to their potential for mechanical reinforcing, osseointegration, and antimicrobial properties. Numerous developments in the fabrication and biological studies of CNMs have provided various opportunities to expand their application to dental regeneration and restoration. In this review, we discuss the advances in novel dental implants with CNMs in terms of tissue engineering, including material combination, coating strategies, and biofunctionalities. We present a brief overview of recent findings and progression in the research to show the promising aspect of CNMs for dental implant application. In conclusion, it is shown that further development of surface functionalization with CNMs may provide innovative results with clinical potential for improved osseointegration after implantation.
Collapse
Affiliation(s)
- Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Korea; (M.S.K.); (H.J.J.); (S.H.L.)
| | - Hee Jeong Jang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Korea; (M.S.K.); (H.J.J.); (S.H.L.)
| | - Seok Hyun Lee
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Korea; (M.S.K.); (H.J.J.); (S.H.L.)
| | - Ji Eun Lee
- Department of Optics and Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Korea; (J.E.L.); (H.J.J.)
| | - Hyo Jung Jo
- Department of Optics and Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Korea; (J.E.L.); (H.J.J.)
| | | | - Bongju Kim
- Dental Life Science Research Institute/Innovation Research & Support Center for Dental Science, Seoul National University Dental Hospital, Seoul 03080, Korea
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Korea; (M.S.K.); (H.J.J.); (S.H.L.)
- Department of Optics and Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Korea; (J.E.L.); (H.J.J.)
| |
Collapse
|
150
|
Suryani L, Foo JKR, Cardilla A, Dong Y, Muthukumaran P, Hassanbhai A, Wen F, Simon DT, Iandolo D, Yu N, Ng KW, Teoh SH. Effects of Pulsed Electromagnetic Field Intensity on Mesenchymal Stem Cells. Bioelectricity 2021. [DOI: 10.1089/bioe.2021.0002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Luvita Suryani
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Jyong Kiat Reuben Foo
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Angelysia Cardilla
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Yibing Dong
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Padmalosini Muthukumaran
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Ammar Hassanbhai
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Feng Wen
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
| | - Daniel T. Simon
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - Donata Iandolo
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
- UMR5510 MATEIS, CNRS, INSA-Lyon, University of Lyon, Lyon, France
- Mines Saint-Etienne, INSERM, U1059 SAINBIOSE, Saint-Étienne, France
| | - Na Yu
- National Dental Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
- Environmental Chemistry & Materials Centre, Nanyang Environment and Water Research Institute (NEWRI), Nanyang Technological University, Singapore, Singapore
- Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Swee-Hin Teoh
- School of Chemical and Biomedical Engineering, College of Engineering, Nanyang Technological University, Singapore, Singapore
- Lee Kong Chian School of Medicine, Singapore, Singapore
| |
Collapse
|