201
|
Zhang B, Huo S, Cen X, Pan X, Huang X, Zhao Z. circAKT3 positively regulates osteogenic differentiation of human dental pulp stromal cells via miR-206/CX43 axis. Stem Cell Res Ther 2020; 11:531. [PMID: 33298186 PMCID: PMC7726914 DOI: 10.1186/s13287-020-02058-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background Human dental pulp stromal cells (hDPSCs) are promising sources of mesenchymal stem cells (MSCs) for bone tissue regeneration. Circular RNAs (circRNAs) have been demonstrated to play critical roles in stem cell osteogenic differentiation. Herein, we aimed to investigate the role of circAKT3 during osteogenesis of hDPSCs and the underlying mechanisms of its function. Methods We performed circRNA sequencing to investigate the expression profiles of circular RNAs during osteogenesis of hDPSCs. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to detect the expression pattern of circAKT3 and miR-206 in hDPSCs during osteogenesis. We knocked down circAKT3 and interfered the expression of miR-206 to verify their regulatory role in hDPSC osteogenesis. We detected hDPSCs mineralization by alkaline phosphatase (ALP) and Alizarin Red S (ARS) staining and used dual-luciferase reporter assay to validate the direct binding between circAKT3 and miR-206. To investigate in vivo mineralization, we performed subcutaneous transplantation in nude mice and used hematoxylin and eosin, Masson’s trichrome, and immunohistochemistry staining. Results Totally, 86 circRNAs were differentially expressed during hDPSC osteogenesis, in which 29 were downregulated while 57 were upregulated. circAKT3 was upregulated while miR-206 was downregulated during hDPSC osteogenesis. Knockdown of circAKT3 inhibited ALP/ARS staining and expression levels of osteogenic genes. circAKT3 directly interacted with miR-206, and the latter one suppressed osteogenesis of hDPSCs. Silencing miR-206 partially reversed the inhibitory effect of circAKT3 knockdown on osteogenesis. Connexin 43 (CX43), which positively regulates osteogenesis of stem cells, was predicted as a target of miR-206, and overexpression or knockdown of miR-206 could correspondingly decrease and increase the expression of CX43. In vivo study showed knockdown of circAKT3 suppressed the formation of mineralized nodules and expression of osteogenic proteins. Conclusion During osteogenesis of hDPSCs, circAKT3 could function as a positive regulator by directly sponging miR-206 and arresting the inhibitive effect of miR-206 on CX43 expression. Supplementary information The online version contains supplementary material available at 10.1186/s13287-020-02058-y.
Collapse
Affiliation(s)
- Bo Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Sibei Huo
- Department of Stomatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders (Chongqing), Chongqing, People's Republic of China
| | - Xiao Cen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Temporomandibular Joint, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Xuefeng Pan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xinqi Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China. .,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China. .,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Section, South Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
202
|
Dorcemus DL, Kim HS, Nukavarapu SP. Gradient scaffold with spatial growth factor profile for osteochondral interface engineering. Biomed Mater 2020; 16. [PMID: 33291092 DOI: 10.1088/1748-605x/abd1ba] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 12/08/2020] [Indexed: 11/11/2022]
Abstract
Osteochondral (OC) matrix design poses a significant engineering challenge due to the complexity involved with bone-cartilage interfaces. To better facilitate the regeneration of OC tissue, we developed and evaluated a biodegradable matrix with uniquely arranged bone and cartilage supporting phases: a poly(lactic-co-glycolic) acid (PLGA) template structure with a porosity gradient along its longitudinal axis uniquely integrated with hyaluronic acid hydrogel. Micro-CT scanning and imaging confirmed the formation of an inverse gradient matrix. Hydroxyapatite was added to the PLGA template which was then plasma-treated to increase hydrophilicity and growth factor affinity. An osteogenic growth factor (bone morphogenetic protein 2; BMP-2) was loaded onto the template scaffold via adsorption, while a chondrogenic growth factor (transforming growth factor beta 1; TGF-β1) was incorporated into the hydrogel phase. Confocal microscopy of the growth factor loaded matrix confirmed the spatial distribution of the two growth factors, with chondrogenic factor confined to the cartilaginous portion and osteogenic factor present throughout the scaffold. We observed spatial differentiation of human mesenchymal stem cells (hMSCs) into cartilage and bone cells in the scaffolds in vitro: cartilaginous regions were marked by increased glycosaminoglycan production, and osteogenesis was seen throughout the graft by alizarin red staining. In a dose-dependent study of BMP-2, hMSC pellet cultures with TGF-β1 and BMP-2 showed synergistic effects on chondrogenesis. These results indicate that development of an inverse gradient matrix can spatially distribute two different growth factors to facilitate chondrogenesis and osteogenesis along different portions of a scaffold, which are key steps needed for formation of an osteochondral interface.
Collapse
Affiliation(s)
- Deborah Leonie Dorcemus
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, Connecticut, 06269, UNITED STATES
| | - Hyun Sung Kim
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, Connecticut, 06269, UNITED STATES
| | - Syam Prasad Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, Connecticut, 06269, UNITED STATES
| |
Collapse
|
203
|
Perczel-Kovách K, Hegedűs O, Földes A, Sangngoen T, Kálló K, Steward MC, Varga G, Nagy KS. STRO-1 positive cell expansion during osteogenic differentiation: A comparative study of three mesenchymal stem cell types of dental origin. Arch Oral Biol 2020; 122:104995. [PMID: 33278647 DOI: 10.1016/j.archoralbio.2020.104995] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/31/2020] [Accepted: 11/16/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Although the osteogenic differentiation potential of mesenchymal stem cells of dental origin is well established, the roles of different marker proteins in this process remain to be clarified. Our aim was to compare the cellular and molecular changes, focusing in particular on mesenchymal stem cell markers, during in vitro osteogenesis in three dental stem cell types: dental follicle stem cells (DFSCs), periodontal ligament stem cells (PDLSCs) and dental pulp stem cells (DPSCs). DESIGN Human DFSCs, PDLSCs and DPSCs were isolated, cultured and their osteogenic differentiation was induced for 3 weeks. Mineralization was assessed by von Kossa staining and calcium concentration measurements. The expression of mesenchymal and osteogenic markers was studied by immunocytochemistry and qPCR techniques. Alkaline phosphatase (ALP) activity and the frequency of STRO-1 positive cells were also quantified. RESULTS The three cultures all showed abundant mineralization, with high calcium content by day 21. The expression of vimentin and nestin was sustained after osteogenic induction. The osteogenic medium induced a considerable elevation of STRO-1 positive cells. By day 7, the ALP mRNA level had increased more than 100-fold in DFSCs, PDLSCs, and DPSCs. Quantitative PCR results indicated dissimilarities of osteoblastic marker levels in the three dental stem cell cultures. CONCLUSIONS DFSCs, PDLSCs and DPSCs have similar functional osteogenic differentiation capacities although their expressional profiles of key osteogenic markers show considerable variations. The STRO-1 positive cell fraction expands during osteogenic differentiation while vimentin and nestin expression remain high. For identification of stemness, functional studies rather than marker expressions are needed.
Collapse
Affiliation(s)
- Katalin Perczel-Kovách
- Department of Oral Biology, Semmelweis University, Nagyvárad Square 4. H-1089 Budapest, Hungary.
| | - Orsolya Hegedűs
- Department of Oral Biology, Semmelweis University, Nagyvárad Square 4. H-1089 Budapest, Hungary.
| | - Anna Földes
- Department of Oral Biology, Semmelweis University, Nagyvárad Square 4. H-1089 Budapest, Hungary.
| | - Thanyaporn Sangngoen
- Department of Oral Biology, Semmelweis University, Nagyvárad Square 4. H-1089 Budapest, Hungary.
| | - Karola Kálló
- Department of Oral Biology, Semmelweis University, Nagyvárad Square 4. H-1089 Budapest, Hungary
| | - Martin C Steward
- Department of Oral Biology, Semmelweis University, Nagyvárad Square 4. H-1089 Budapest, Hungary; School of Medical Sciences, University of Manchester, Manchester, United Kingdom.
| | - Gábor Varga
- Department of Oral Biology, Semmelweis University, Nagyvárad Square 4. H-1089 Budapest, Hungary.
| | - Krisztina S Nagy
- Department of Oral Biology, Semmelweis University, Nagyvárad Square 4. H-1089 Budapest, Hungary.
| |
Collapse
|
204
|
Zhang Q, Zuo H, Yu S, Lin Y, Chen S, Liu H, Chen Z. RUNX2 co-operates with EGR1 to regulate osteogenic differentiation through Htra1 enhancers. J Cell Physiol 2020; 235:8601-8612. [PMID: 32324256 PMCID: PMC8895429 DOI: 10.1002/jcp.29704] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/20/2020] [Accepted: 03/31/2020] [Indexed: 11/19/2023]
Abstract
Runt-related transcription factor 2 (Runx2) has been shown to regulate osteoblast differentiation by directly or indirectly regulating numerous osteoblast-related genes. However, our understanding of the transcriptional mechanisms of RUNX2 is mainly restricted to its transactivation, while the mechanism underlying its inhibitory effect during osteoblast differentiation remains largely unknown. Here, we incorporated the anti-RUNX2 chromatin immunoprecipitation (ChIP) sequencing in MC3T3-E1 cells and RNA-sequencing of parietal bone from Runx2 heterozygous mutant mice, to identify the putative genes negatively regulated by RUNX2. We identified HtrA serine peptidase 1 (Htra1) as a target gene and found ten candidate Htra1 enhancers potentially regulated by RUNX2, among which seven were verified by dual-luciferase assays. Furthermore, we investigated the motifs in the vicinity of RUNX2-binding sites and identified early growth response 1 (EGR1) as a potential partner transcription factor (TF) potentially regulating Htra1 expression, which was subsequently confirmed by Re-ChIP assays. RUNX2 and EGR1 co-repressed Htra1 and increased the expression levels of other osteoblast marker genes, such as osterix, osteocalcin, and osteoprotegerin at the messenger RNA and protein level. Moreover, Alizarin red staining combined with alkaline phosphatase (ALP) staining showed decreased calcified nodules and ALP activity in the siRUNX2+siEGR1 group compared with siRUNX2 group. Our findings revealed the detailed mechanism of the inhibitory function of RUNX2 towards its downstream genes, along with its partner TFs, to promote osteoblast differentiation.
Collapse
Affiliation(s)
- Qian Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Huanyan Zuo
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shuaitong Yu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yuxiu Lin
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shuo Chen
- Department of Developmental Dentistry, University of Texas Health Science Center, San Antonio, Texas
| | - Huan Liu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi Chen
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
205
|
Concentrated Growth Factors (CGF) Induce Osteogenic Differentiation in Human Bone Marrow Stem Cells. BIOLOGY 2020; 9:biology9110370. [PMID: 33143015 PMCID: PMC7693660 DOI: 10.3390/biology9110370] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022]
Abstract
Bone regeneration is a complex process regulated by several factors that control overlapping biological processes, coordinating interactions among distinct cell populations. There is a great interest in identifying new strategies for inducing osteogenesis in a safe and efficient manner. Concentrated Growth Factor (CGF) is an autologous blood derived product obtained by centrifugation of venous blood following the procedure set on the Silfradent device. In this study the effects of CGF on osteogenic differentiation of human Bone Marrow Stem Cells (hBMSC) in vitro have been investigated; hBMSC were cultured with CGF or osteogenic medium, for 21 days. The osteogenic differentiation was evaluated measuring alkaline phosphatase (ALP) enzyme activity, matrix mineralization by alizarin red staining and through mRNA and protein quantification of osteogenic differentiation markers by Real-time PCR and Western blotting, respectively. The treatment with CGF stimulated ALP activity and promoted matrix mineralization compared to control and seems to be more effective than osteogenic medium. Also, hBMSC lost mesenchymal markers and showed other osteogenic features. Our study showed for the first time that CGF alone is able to induce osteogenic differentiation in hBMSC. The application of CGF on hBMSC osteoinduction might offer new clinical and biotechnological strategies in the tissue regeneration field.
Collapse
|
206
|
de Melo Pereira D, Eischen-Loges M, Birgani ZT, Habibovic P. Proliferation and Osteogenic Differentiation of hMSCs on Biomineralized Collagen. Front Bioeng Biotechnol 2020; 8:554565. [PMID: 33195119 PMCID: PMC7644787 DOI: 10.3389/fbioe.2020.554565] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/05/2020] [Indexed: 01/12/2023] Open
Abstract
Biomineralized collagen with intrafibrillar calcium phosphate mineral provides an excellent mimic of the composition and structure of the extracellular matrix of bone, from nano- to micro-scale. Scaffolds prepared from this material have the potential to become the next-generation of synthetic bone graft substitutes, as their unique properties make them closer to the native tissue than synthetic alternatives currently available to clinicians. To understand the interaction between biomineralized collagen and cells that are relevant in the context of bone regeneration, we studied the growth and osteogenic differentiation of bone marrow derived human mesenchymal stromal cells (hMSCs) cultured on biomineralized collagen membranes, and compared it to the cell behavior on collagen membranes without mineral. Cells proliferated normally on both biomimetic membranes, and were more triggered to differentiate toward the osteogenic lineage by the biomineralized collagen. This was shown by the elevated mRNA levels of RUNX2, SPP1, ENPP1, and OCN after 3 days of culture, and COL1A1 after 14 days of culture on mineralized collagen. The mRNA levels of the tested markers of osteogenesis were lower on collagen membranes without mineral, with the exception of OCN, which was more highly expressed on collagen than on biomineralized collagen membranes. Expression by hMSCs of OPG, a gene involved in inhibition of osteoclastogenesis, was higher on biomineralized collagen at day 3, while M-CSF, involved in osteoblast-osteoclast communication, was upregulated on both membranes at day 3 and 14 of culture. Alkaline phosphatase activity of hMSCs was high on both biomimetic membranes when compared with cells cultured on tissue culture plastic. Cell-induced mineralization was observed on collagen membranes, while the high mineral content of the biomineralized membranes prohibited a reliable analysis of cell-induced mineralization on these membranes. In conclusion, we have identified that both collagen and biomineralized collagen support proliferation, osteogenic differentiation and mineralization of hMSCs, with biomineralized membranes having a more pronounced positive effect. These findings support the existing evidence that biomineralized collagen is a promising material in the field of bone regeneration.
Collapse
Affiliation(s)
| | | | | | - Pamela Habibovic
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
207
|
West-Livingston LN, Park J, Lee SJ, Atala A, Yoo JJ. The Role of the Microenvironment in Controlling the Fate of Bioprinted Stem Cells. Chem Rev 2020; 120:11056-11092. [PMID: 32558555 PMCID: PMC7676498 DOI: 10.1021/acs.chemrev.0c00126] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The field of tissue engineering and regenerative medicine has made numerous advances in recent years in the arena of fabricating multifunctional, three-dimensional (3D) tissue constructs. This can be attributed to novel approaches in the bioprinting of stem cells. There are expansive options in bioprinting technology that have become more refined and specialized over the years, and stem cells address many limitations in cell source, expansion, and development of bioengineered tissue constructs. While bioprinted stem cells present an opportunity to replicate physiological microenvironments with precision, the future of this practice relies heavily on the optimization of the cellular microenvironment. To fabricate tissue constructs that are useful in replicating physiological conditions in laboratory settings, or in preparation for transplantation to a living host, the microenvironment must mimic conditions that allow bioprinted stem cells to proliferate, differentiate, and migrate. The advances of bioprinting stem cells and directing cell fate have the potential to provide feasible and translatable approach to creating complex tissues and organs. This review will examine the methods through which bioprinted stem cells are differentiated into desired cell lineages through biochemical, biological, and biomechanical techniques.
Collapse
Affiliation(s)
- Lauren N. West-Livingston
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Jihoon Park
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - James J. Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|
208
|
Nanoparticle-directed and ionically forced polyphosphate coacervation: a versatile and reversible core-shell system for drug delivery. Sci Rep 2020; 10:17147. [PMID: 33051468 PMCID: PMC7555899 DOI: 10.1038/s41598-020-73100-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/10/2020] [Indexed: 12/11/2022] Open
Abstract
A drug encapsulation/delivery system using a novel principle is described that is based on an intra-particle migration of calcium ions between a central Ca2+-enriched nanoparticle core and the surrounding shell compartment. The supply of Ca2+ is needed for the formation of a coacervate shell around the nanoparticles, acting as the core of drug-loadable core–shell particles, using the physiological inorganic polymer polyphosphate (polyP). This polyanion has the unique property to form, at an alkaline pH and in the presence of a stoichiometric surplus of calcium ions, water-insoluble and stabile amorphous nanoparticles. At neutral pH a coacervate, the biologically active form of the polymer, is obtained that is composed of polyP and Ca2+. The drug-loaded core–shell particles, built from the Ca–polyP core and the surrounding Ca–polyP shell, were fabricated in two successive steps. First, the formation of the nanoparticle core at pH 10 and a superstoichiometric 2:1 molar ratio between CaCl2 and Na–polyP into which dexamethasone, as a phosphate derivative, was incorporated. Second, the preparation of the coacervate shell, loaded with ascorbic acid, by exposure of the Ca–polyP core to soluble Na–polyP and L-ascorbate (calcium salt). EDX analysis revealed that during this step the Ca2+ ions required for coacervate formation migrate from the Ca–polyP core (with a high Ca:P ratio) to the shell. Electron microscopy of the particles show an electron-dense 150–200 nm sized core surrounded by a less sharply delimited electron-sparse shell. The core–shell particles exhibited strong osteogenic activity in vitro, based on the combined action of polyP and of dexamethasone and ascorbic acid, which reversibly bind to the anionic polyP via ionic Ca2+ bonds. Drug release from the particles occurs after contact with a peptide/protein-containing serum, a process which is almost complete after 10 days and accompanied by the conversion of the nanoparticles into a coacervate. Human osteosarcoma SaOS-2 cells cultivated onto or within an alginate hydrogel matrix showed increased growth/viability and mineralization when the hybrid particles containing dexamethasone and ascorbic acid were embedded in the matrix. The polyP-based core–shell particles have the potential to become a suitable, pH-responsive drug encapsulation/release system, especially for bone, cartilage and wound healing.
Collapse
|
209
|
Madruga LYC, Balaban RC, Popat KC, Kipper MJ. Biocompatible Crosslinked Nanofibers of Poly(Vinyl Alcohol)/Carboxymethyl-Kappa-Carrageenan Produced by a Green Process. Macromol Biosci 2020; 21:e2000292. [PMID: 33021064 DOI: 10.1002/mabi.202000292] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/19/2020] [Indexed: 12/26/2022]
Abstract
This study presents a new type of biocompatible nanofiber based on poly(vinyl alcohol) (PVA) and carboxymethyl-kappa-carrageenan (CMKC) blends, produced with no generation of hazardous waste. The nanofibers are produced by electrospinning using PVA:CMKC blends with ratios of 1:0, 1:0.25, 1:0.4, 1:0.5, and 1:0.75 (w/w PVA:CMKC) in aqueous solution, followed by thermal crosslinking. The diameter of the fibers is in the nanometer scale and below 300 nm. Fourier transform infrared spectroscopy shows the presence of the carboxyl and sulfate groups in all the fibers with CMKC. The nanofibers from water-soluble polymers are stabilized by thermal crosslinking. The incorporation of CMKC improves cytocompatibility, biodegradability, cell growth, and cell adhesion, compared to PVA nanofibers. Furthermore, the incorporation of CMKC modulates phenotype of human adipose-derived stem cells (ADSCs). PVA/CMKC nanofibers enhance ADSC response to osteogenic differentiation signals and are therefore good candidates for application in tissue engineering to support stem cells.
Collapse
Affiliation(s)
- Liszt Y C Madruga
- Institute of Chemistry, Federal University of Rio Grande do Norte (UFRN), Natal, RN, 59078-970, Brazil.,Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Rosangela C Balaban
- Institute of Chemistry, Federal University of Rio Grande do Norte (UFRN), Natal, RN, 59078-970, Brazil
| | - Ketul C Popat
- School of Advanced Materials Discovery, Colorado State University, Fort Collins, CO, 80523, USA.,School of Biomedical Engineering, Colorado State University, Fort Collins, CO, 80523, USA.,Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Matt J Kipper
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, 80523, USA.,School of Advanced Materials Discovery, Colorado State University, Fort Collins, CO, 80523, USA.,School of Biomedical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| |
Collapse
|
210
|
E LL, Cheng T, Li CJ, Zhang R, Zhang S, Liu HC, Zheng WJ. Combined Use of Recombinant Human BMP-7 and Osteogenic Media May Have No Ideal Synergistic Effect on Leporine Bone Regeneration of Human Umbilical Cord Mesenchymal Stem Cells Seeded on Nanohydroxyapatite/Collagen/Poly (l-Lactide). Stem Cells Dev 2020; 29:1215-1228. [PMID: 32674666 DOI: 10.1089/scd.2020.0066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Human umbilical cord mesenchymal stem cells (hUC-MSCs) are a promising alternative source of mesenchymal stem cells (MSCs) that are enormously attractive for clinical use. This study was designed to investigate the effect of recombinant human bone morphogenetic protein-7 (rhBMP-7) and/or osteogenic media (OMD) on bone regeneration of hUC-MSCs seeded on nanohydroxyapatite/collagen/poly(l-lactide) (nHAC/PLA) in a rabbit model. The characteristics of stem cells were analyzed by plastic adherence, cell phenotype, and multilineage differentiation potential. Cell proliferation was examined using cell counting kit-8 assay. Osteogenic differentiation was evaluated by quantitative Ca2+ concentration, PO43- concentration, alkaline phosphatase (ALP) activity, osteocalcin (OCN) secretion, and mineralized matrix formation. Bone regeneration was investigated in jaw bone defect repair in rabbit by microcomputed tomography, fluorescent labeling, and hematoxylin and eosin staining. Except for initial stress response, OMD and OMD + rhBMP-7 inhibited the proliferation of hUC-MSCs seeded on nHAC/PLA; rhBMP-7 inhibited cell proliferation in the nonlogarithmic phase and attenuated the inhibitory effect of OMD on cell proliferation. The inhibitory effects of OMD, rhBMP-7, and OMD + rhBMP-7 on cell proliferation were ranked as OMD > OMD + rhBMP-7 > rhBMP-7. OMD, rhBMP-7, and OMD + rhBMP-7 promoted Ca2+ concentration, PO43- concentration, ALP activity, OCN secretion, and mineralized matrix formation of hUC-MSCs seeded on nHAC/PLA. The promoting effects of OMD, rhBMP-7, and OMD+rhBMP-7 on Ca2+ concentration, PO43- concentration, ALP activity, OCN secretion, and mineralized matrix formation were ranked as rhBMP-7 > OMD > OMD + rhBMP-7, OMD > OMD + rhBMP-7 > rhBMP-7, OMD > rhBMP-7 > OMD + rhBMP-7, rhBMP-7 > OMD + rhBMP-7 > OMD, and OMD > rhBMP-7 > OMD + rhBMP-7, respectively. In rabbit jaw bone defect repair, OMD, rhBMP-7, and OMD + rhBMP-7 enhanced bone regeneration of hUC-MSCs seeded on nHAC/PLA, but the largest bone mineral apposition rate and bone formation were presented in cultures with rhBMP-7. These findings suggested that the combined use of rhBMP-7 and OMD may have no ideal synergistic effect on bone regeneration of hUC-MSCs seeded on nHAC/PLA in rabbit jaw bone defect.
Collapse
Affiliation(s)
- Ling-Ling E
- Department of Chemistry, Jinan University, Guangzhou, China.,Institute of Stomatology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Tao Cheng
- Institute of Stomatology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Chuan-Jie Li
- Institute of Stomatology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Rong Zhang
- Institute of Stomatology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Shuo Zhang
- Institute of Stomatology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hong-Chen Liu
- Institute of Stomatology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Wen-Jie Zheng
- Department of Chemistry, Jinan University, Guangzhou, China
| |
Collapse
|
211
|
Soheilmoghaddam M, Padmanabhan H, Cooper-White JJ. Biomimetic cues from poly(lactic-co-glycolic acid)/hydroxyapatite nano-fibrous scaffolds drive osteogenic commitment in human mesenchymal stem cells in the absence of osteogenic factor supplements. Biomater Sci 2020; 8:5677-5689. [PMID: 32915185 DOI: 10.1039/d0bm00946f] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mimicking the complex hierarchical architecture of the 'osteon', the functional unit of cortical bone, from the bottom-up offers the possibility of generating mature bone tissue in tissue engineered bone substitutes. In this work, a modular 'bottom-up' approach has been developed to assemble bone niche-mimicking nanocomposite scaffolds composed of aligned electrospun nanofibers of poly(lactic-co-glycolic acid) (PLGA) encapsulating aligned rod-shape nano-sized hydroxyapatite (nHA). By encoding axial orientation of the nHA within these aligned nanocomposite fibers, significant improvements in mechanical properties, surface roughness, hydrophilicity and in vitro simulated body fluid (SBF) mineral deposition were achieved. Moreover, these hierarchical scaffolds induced robust formation of bone hydroxyapatite and osteoblastic maturation of human bone marrow-derived mesenchymal stem cells (hBMSCs) in growth media that was absent of any soluble osteogenic differentiation factors. The results of this investigation confirm that these tailored, aligned nanocomposite fibers, in the absence of media-bone inductive factors, offer the requisite biophysical and biochemical cues to hBMSCs to promote and support their differentiation into mature osteoblast cells and form early bone-like tissue in vitro.
Collapse
Affiliation(s)
- Mohammad Soheilmoghaddam
- Tissue Engineering and Microfluidics Laboratory (TE&M), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St Lucia, QLD, Australia.
| | | | | |
Collapse
|
212
|
Upadhyai P, Guleria VS, Udupa P. Characterization of primary cilia features reveal cell-type specific variability in in vitro models of osteogenic and chondrogenic differentiation. PeerJ 2020; 8:e9799. [PMID: 32884864 PMCID: PMC7444507 DOI: 10.7717/peerj.9799] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Primary cilia are non-motile sensory antennae present on most vertebrate cell surfaces. They serve to transduce and integrate diverse external stimuli into functional cellular responses vital for development, differentiation and homeostasis. Ciliary characteristics, such as length, structure and frequency are often tailored to distinct differentiated cell states. Primary cilia are present on a variety of skeletal cell-types and facilitate the assimilation of sensory cues to direct skeletal development and repair. However, there is limited knowledge of ciliary variation in response to the activation of distinct differentiation cascades in different skeletal cell-types. C3H10T1/2, MC3T3-E1 and ATDC5 cells are mesenchymal stem cells, preosteoblast and prechondrocyte cell-lines, respectively. They are commonly employed in numerous in vitro studies, investigating the molecular mechanisms underlying osteoblast and chondrocyte differentiation, skeletal disease and repair. Here we sought to evaluate the primary cilia length and frequencies during osteogenic differentiation in C3H10T1/2 and MC3T3-E1 and chondrogenic differentiation in ATDC5 cells, over a period of 21 days. Our data inform on the presence of stable cilia to orchestrate signaling and dynamic alterations in their features during extended periods of differentiation. Taken together with existing literature these findings reflect the occurrence of not only lineage but cell-type specific variation in ciliary attributes during differentiation. These results extend our current knowledge, shining light on the variabilities in primary cilia features correlated with distinct differentiated cell phenotypes. It may have broader implications in studies using these cell-lines to explore cilia dependent cellular processes and treatment modalities for skeletal disorders centered on cilia modulation.
Collapse
Affiliation(s)
- Priyanka Upadhyai
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Vishal Singh Guleria
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Prajna Udupa
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
213
|
Lee H, Kim SHL, Yoon H, Ryu J, Park HH, Hwang NS, Park TH. Intracellular Delivery of Recombinant RUNX2 Facilitated by Cell-Penetrating Protein for the Osteogenic Differentiation of hMSCs. ACS Biomater Sci Eng 2020; 6:5202-5214. [PMID: 33455270 DOI: 10.1021/acsbiomaterials.0c00827] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are a commonly used cell source for cell therapy and tissue engineering because of their easy accessibility and multipotency. Runt-related transcription factor 2 (RUNX2) is a master regulator of the osteogenic commitment of hMSCs. Either recombinant plasmid delivery or viral transduction has been utilized to activate RUNX2 gene expression for effective hMSC differentiation. In this study, recombinant RUNX2 fused with cell-penetrating 30Kc19α protein (30Kc19α-RUNX2) was delivered into hMSCs for osteogenic commitment. Fusion of recombinant RUNX2 with 30Kc19α resulted in successful delivery of the protein into cells and enhanced soluble expression of the protein. Intracellular delivery of the 30Kc19α-RUNX2 fusion protein enhanced the osteogenic differentiation of hMSCs in vitro. 30Kc19α-RUNX2 treatment resulted in increased ALP accumulation and elevated calcium deposition. Finally, implantation of hMSCs treated with 30Kc19α-RUNX2 showed osteogenesis via cell delivery into the subcutaneous tissue and bone regeneration in a cranial defect mouse model. Therefore, we suggest that 30Kc19α-RUNX2, an osteoinductive recombinant protein, is an efficient tool for bone tissue engineering.
Collapse
Affiliation(s)
- Haein Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung Hyun L Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyungro Yoon
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jina Ryu
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Hee Ho Park
- Department of Biotechnology and Bioengineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.,Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea.,BioMax/N-Bio Institute, Institute of Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.,Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea.,BioMax/N-Bio Institute, Institute of Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
214
|
circRNA Expression Profile in Dental Pulp Stem Cells during Odontogenic Differentiation. Stem Cells Int 2020; 2020:5405931. [PMID: 32952566 PMCID: PMC7482017 DOI: 10.1155/2020/5405931] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 06/25/2020] [Accepted: 08/08/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction Odontogenic differentiation of human dental pulp stem cells (hDPSCs) is a key step of pulp regeneration. Recent studies showed that circular RNAs (circRNAs) have many biological functions and that competing endogenous RNA (ceRNA) is their most common mechanism of action. However, the role of circRNAs in hDPSCs during odontogenesis is still unclear. Methods Isolated hDPSCs were cultured in essential and odontogenic medium. Total RNA was extracted after 14 days of culture, and then, microarray analysis was performed to measure the differential expressions of circRNAs. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was then performed to validate the microarray results. Based on microarray data from this study and available in the database, a ceRNA network was constructed to investigate the potential function of circRNAs during odontogenesis. In addition, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to investigate the potential correlation between signaling pathways and circRNAs. In addition, qRT-PCR and Western blot analysis were used to explore the function of hsa_circRNA_104101. Results We found 43 upregulated circRNAs and 144 downregulated circRNAs during the odontogenic differentiation process (fold change > 1.5 and <-1.5, respectively; P < 0.05). qRT-PCR results were in agreement with the microarray results. Bioinformatic analysis revealed that the Wnt signaling pathway and the TGF-β signaling pathway, as well as the other pathways associated with odontogenic differentiation, were correlated to the differentially expressed circRNAs. hsa_circRNA_104101 was proved to promote the odontogenic differentiation of hDPSCs. Conclusion This study reported 187 circRNAs that were differentially expressed in hDPSCs during odontogenic differentiation. Bioinformatic analysis of the expression data suggested that circRNA-miRNA-mRNA networks might act as a crucial mechanism for hDPSC odontogenic differentiation, providing a theoretical foundation for the study of pulp regeneration regulation by circRNAs.
Collapse
|
215
|
Dong J, Xu X, Zhang Q, Yuan Z, Tan B. Dkk1 acts as a negative regulator in the osteogenic differentiation of the posterior longitudinal ligament cells. Cell Biol Int 2020; 44:2450-2458. [PMID: 32827333 DOI: 10.1002/cbin.11452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/22/2020] [Accepted: 08/16/2020] [Indexed: 12/13/2022]
Abstract
Ossification of the posterior longitudinal ligament (OPLL) is a spinal disorder characterized by progressive ectopic bone formation in the PLL of the spine. Dickkopf-1 (Dkk1) is a secreted inhibitor of the Wnt pathway that negatively regulates bone formation during skeletal development. However, whether Dkk1 impacts the pathogenesis of OPLL has not been reported. This study is to investigate the role of Dkk1 in the development of OPLL. Our results show that the serum levels of Dkk1 are decreased in OPLL patients compared with non-OPLL controls. The expression of Dkk1 is also reduced in OPLL ligament cells. Downregulation of Dkk1 in ligament cells is associated with activation of the Wnt/β-catenin signaling, as indicated by stabilized β-catenin and increased T-cell factor-dependent transcriptional activity. Functionally, Dkk1 exerts a growth-inhibitory effect by repressing proliferation but promoting apoptosis of ligament cells. Dkk1 also suppresses bone morphogenetic protein 2-induced entire osteogenic differentiation of ligament cells, and this suppression is mediated via its inhibition of the Wnt pathway. Our results demonstrate for the first time that Dkk1 acts as an important negative regulator in the ossification of the PLL. Targeting the Wnt pathway using Dkk1 may represent a potential therapeutic strategy for the treatment of OPLL.
Collapse
Affiliation(s)
- Jun Dong
- Department of Orthopaedics, Shangdong Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiqiang Xu
- Department of Orthopaedics, Shangdong Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qingyu Zhang
- Department of Orthopaedics, Shangdong Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zenong Yuan
- Department of Orthopaedics, Shangdong Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Bingyi Tan
- Department of Orthopaedics, Shangdong Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
216
|
Jiang M, Liu R, Liu L, Kot A, Liu X, Xiao W, Jia J, Li Y, Lam KS, Yao W. Identification of osteogenic progenitor cell-targeted peptides that augment bone formation. Nat Commun 2020; 11:4278. [PMID: 32855388 PMCID: PMC7453024 DOI: 10.1038/s41467-020-17417-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 06/16/2020] [Indexed: 12/02/2022] Open
Abstract
Activation and migration of endogenous mesenchymal stromal cells (MSCs) are critical for bone regeneration. Here, we report a combinational peptide screening strategy for rapid discovery of ligands that not only bind strongly to osteogenic progenitor cells (OPCs) but also stimulate osteogenic cell Akt signaling in those OPCs. Two lead compounds are discovered, YLL3 and YLL8, both of which increase osteoprogenitor osteogenic differentiation in vitro. When given to normal or osteopenic mice, the compounds increase mineral apposition rate, bone formation, bone mass, and bone strength, as well as expedite fracture repair through stimulated endogenous osteogenesis. When covalently conjugated to alendronate, YLLs acquire an additional function resulting in a “tri-functional” compound that: (i) binds to OPCs, (ii) targets bone, and (iii) induces “pro-survival” signal. These bone-targeted, osteogenic peptides are well suited for current tissue-specific therapeutic paradigms to augment the endogenous osteogenic cells for bone regeneration and the treatment of bone loss. Activation of osteogenic cells is essential for bone regeneration. Here, the authors screen a peptide library and identify 2 compounds that promote osteogenic progenitor cell differentiation in vitro, and show that they increase bone formation and fracture repair in mice.
Collapse
Affiliation(s)
- Min Jiang
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA.,Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025, Shanghai, China
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Lixian Liu
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Alexander Kot
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Xueping Liu
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Junjing Jia
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Wei Yao
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
217
|
Vis MAM, Ito K, Hofmann S. Impact of Culture Medium on Cellular Interactions in in vitro Co-culture Systems. Front Bioeng Biotechnol 2020; 8:911. [PMID: 32850750 PMCID: PMC7417654 DOI: 10.3389/fbioe.2020.00911] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/15/2020] [Indexed: 12/24/2022] Open
Abstract
Co-culturing of cells in in vitro tissue models is widely used to study how they interact with each other. These models serve to represent a variety of processes in the human body such as development, homeostasis, regeneration, and disease. The success of a co-culture is dependent on a large number of factors which makes it a complex and ambiguous task. This review article addresses co-culturing challenges regarding the cell culture medium used in these models, in particular concerning medium composition, volume, and exchange. The effect of medium exchange on cells is often an overlooked topic but particularly important when cell communication via soluble factors and extracellular vesicles, the so-called cell secretome (CS) is being studied. Culture medium is regularly exchanged to supply new nutrients and to eliminate waste products produced by the cells. By removing medium, important CSs are also removed. After every medium change, the cells must thus restore their auto- and paracrine communication through these CSs. This review article will also discuss the possibility to integrate biosensors into co-cultures, in particular to provide real-time information regarding media composition. Overall, the manner in which culture medium is currently used will be re-evaluated. Provided examples will be on the subject of bone tissue engineering.
Collapse
Affiliation(s)
- Michelle A M Vis
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
218
|
Zhao C, Chen Q, Yu S, Xu C, Li X, Zhang C, Gao L. Effect of interleukin-22 on osteogenic differentiation and the osteoclastogenic response of human periodontal ligament fibroblasts in vitro. J Periodontol 2020; 91:1085-1097. [PMID: 31950496 DOI: 10.1002/jper.19-0470] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/06/2019] [Accepted: 11/17/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Interleukin-22 (IL-22) exerts extensive biological effects, playing both protective and pathological roles in autoimmune and infectious diseases. However, the specific role and mechanism of IL-22 in the pathogenesis of periodontitis have not been clarified. The aim of this study was to analyze the possible roles of IL-22 in the osteoclastogenesis and osteogenesis of periodontitis. METHODS Human periodontal ligament fibroblasts (hPDLFs) were treated with IL-22 and/or lipopolysaccharide from Porphyromonas gingivalis (Pg-LPS), and the mRNA and protein expression of RANKL and OPG were measured by qRT-PCR and Western blotting, respectively. Western blotting was also used to examine the phosphorylated and total protein expression of MAPK signaling molecules. The role of the MAPK pathway in osteoclastogenesis marker expression was further confirmed by inhibition assays. For osteogenic assays, the mRNA expression of osteoblastic markers was quantified by qRT-PCR, the alkaline phosphatase (ALP) activity of hPDLFs was measured by an ALP assay, and the mineralized nodules formed by hPDLFs were determined by Alizarin Red S staining. RESULTS IL-22 promoted the expression of RANKL in hPDLFs via the MAPK signaling pathway and further upregulated RANKL expression together with Pg-LPS via the p38 MAPK pathway. IL-22 could enhance the ALP activity and mineralized nodule formation of hPDLFs in the early period of osteogenic induction, while exhibiting no profound effect on the expression of osteoblastic markers. CONCLUSION IL-22 plays regulatory roles in bone homeostasis, and it is likely to contribute to osteoclastogenesis as a proinflammatory cytokine in the pathogenesis of periodontitis.
Collapse
Affiliation(s)
- Chuanjiang Zhao
- Department of Periodontology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qianying Chen
- Department of Periodontology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Shaojie Yu
- Department of Periodontology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chenrong Xu
- Department of Periodontology, Guangdong Provincial Hospital of Stomatology, Stomatological Hospital of Southern Medical University, Guangzhou, China
| | - Xiting Li
- Department of Periodontology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chi Zhang
- Department of Periodontology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Li Gao
- Department of Periodontology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
219
|
Kirsch M, Herder AC, Boudot C, Karau A, Rach J, Handke W, Seltsam A, Scheper T, Lavrentieva A. Xeno-Free In Vitro Cultivation and Osteogenic Differentiation of hAD-MSCs on Resorbable 3D Printed RESOMER ®. MATERIALS 2020; 13:ma13153399. [PMID: 32752065 PMCID: PMC7436127 DOI: 10.3390/ma13153399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/23/2020] [Accepted: 07/28/2020] [Indexed: 01/14/2023]
Abstract
The development of alloplastic resorbable materials can revolutionize the field of implantation technology in regenerative medicine. Additional opportunities to colonize the three-dimensionally (3D) printed constructs with the patient’s own cells prior to implantation can improve the regeneration process but requires optimization of cultivation protocols. Human platelet lysate (hPL) has already proven to be a suitable replacement for fetal calf serum (FCS) in 2D and 3D cell cultures. In this study, we investigated the in vitro biocompatibility of the printed RESOMER® Filament LG D1.75 materials as well as the osteogenic differentiation of human mesenchymal stem cells (hMSCs) cultivated on 3D printed constructs under the influence of different medium supplements (FCS, human serum (HS) and hPL). Additionally, the in vitro degradation of the material was studied over six months. We demonstrated that LG D1.75 is biocompatible and has no in vitro cytotoxic effects on hMSCs. Furthermore, hMSCs grown on the constructs could be differentiated into osteoblasts, especially supported by supplementation with hPL. Over six months under physiological in vitro conditions, a distinct degradation was observed, which, however, had no influence on the biocompatibility of the material. Thus, the overall suitability of the material LG D1.75 to produce 3D printed, resorbable bone implants and the promising use of hPL in the xeno-free cultivation of human MSCs on such implants for autologous transplantation have been demonstrated.
Collapse
Affiliation(s)
- Marline Kirsch
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany; (M.K.); (A.-C.H.); (T.S.)
| | - Annabelle-Christin Herder
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany; (M.K.); (A.-C.H.); (T.S.)
| | - Cécile Boudot
- Evonik Nutrition & Care GmbH, Business Line Health Care, Kirschenallee, 64293 Darmstadt, Germany; (C.B.); (A.K.)
| | - Andreas Karau
- Evonik Nutrition & Care GmbH, Business Line Health Care, Kirschenallee, 64293 Darmstadt, Germany; (C.B.); (A.K.)
| | - Jessica Rach
- German Red Cross Blood Service NSTOB, Institute Springe, Eldagsener Straße 38, 31830 Springe, Germany;
| | - Wiebke Handke
- Bavarian Red Cross Blood Service, Institute Nuremberg, Heimerichstrasse 57, 90419 Nuremberg, Germany; (W.H.); (A.S.)
| | - Axel Seltsam
- Bavarian Red Cross Blood Service, Institute Nuremberg, Heimerichstrasse 57, 90419 Nuremberg, Germany; (W.H.); (A.S.)
| | - Thomas Scheper
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany; (M.K.); (A.-C.H.); (T.S.)
| | - Antonina Lavrentieva
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany; (M.K.); (A.-C.H.); (T.S.)
- Correspondence: ; Tel.: +49-511-762-2955
| |
Collapse
|
220
|
Fushimi A, Takeyama H, Tachibana T, Manome Y. Osteogenic cocktail induces calcifications in human breast cancer cell line via placental alkaline phosphatase expression. Sci Rep 2020; 10:12669. [PMID: 32728117 PMCID: PMC7391638 DOI: 10.1038/s41598-020-69622-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/14/2020] [Indexed: 01/27/2023] Open
Abstract
Breast cancer is frequently characterized by calcifications in mammography. The mechanism for calcifications in breast cancer is not completely known. Understanding this mechanism will improve diagnostic accuracy. Herein, we demonstrated that calcifications occur and that alkaline phosphatase enzyme activity increases in MDA-MB-231 cells cultured using an osteogenic cocktail-containing medium. Microarray transcript analysis showed that the PI3K-Akt signaling pathway was significantly involved, with recruitment of placental alkaline phosphatase. Calcifications and alkaline phosphatase enzyme activity were suppressed by silencing placental alkaline phosphatase using a small interfering RNA. Inhibition of the PI3K-Akt signaling pathway suppressed phospho-c-Jun and placental alkaline phosphatase and resulted in absence of calcifications. These findings reveal that breast cancer cells acquire alkaline phosphatase enzyme activity via placental alkaline phosphatase expression and suggest that breast calcification formation is closely associated with the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Atsushi Fushimi
- Core Research Facilities, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, Japan. .,Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan.
| | - Hiroshi Takeyama
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Toshiaki Tachibana
- Core Research Facilities, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, Japan
| | - Yoshinobu Manome
- Core Research Facilities, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo, Japan
| |
Collapse
|
221
|
Nyambat B, Manga YB, Chen CH, Gankhuyag U, Pratomo WP A, Kumar Satapathy M, Chuang EY. New Insight into Natural Extracellular Matrix: Genipin Cross-Linked Adipose-Derived Stem Cell Extracellular Matrix Gel for Tissue Engineering. Int J Mol Sci 2020; 21:E4864. [PMID: 32660134 PMCID: PMC7402347 DOI: 10.3390/ijms21144864] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 07/01/2020] [Indexed: 01/04/2023] Open
Abstract
The cell-derived extracellular matrix (ECM) is associated with a lower risk of pathogen transfer, and it possesses an ideal niche with growth factors and complex fibrillar proteins for cell attachment and growth. However, the cell-derived ECM is found to have poor biomechanical properties, and processing of cell-derived ECM into gels is scarcely studied. The gel provides platforms for three-dimensional cell culture, as well as injectable biomaterials, which could be delivered via a minimally invasive procedure. Thus, in this study, an adipose-derived stem cell (ADSC)-derived ECM gel was developed and cross-linked by genipin to address the aforementioned issue. The genipin cross-linked ADSC ECM gel was fabricated via several steps, including rabbit ADSC culture, cell sheets, decellularization, freeze-thawing, enzymatic digestion, neutralization of pH, and cross-linking. The physicochemical characteristics and cytocompatibility of the gel were evaluated. The results demonstrated that the genipin cross-linking could significantly enhance the mechanical properties of the ADSC ECM gel. Furthermore, the ADSC ECM was found to contain collagen, fibronectin, biglycan, and transforming growth factor (TGF)-β1, which could substantially maintain ADSC, skin, and ligament fibroblast cell proliferation. This cell-derived natural material could be suitable for future regenerative medicine and tissue engineering application.
Collapse
Affiliation(s)
- Batzaya Nyambat
- Graduate Institute of Biomedical Materials and Tissue Engineering, School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (B.N.); (Y.B.M.); (U.G.); (M.K.S.)
| | - Yankuba B. Manga
- Graduate Institute of Biomedical Materials and Tissue Engineering, School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (B.N.); (Y.B.M.); (U.G.); (M.K.S.)
| | - Chih-Hwa Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (B.N.); (Y.B.M.); (U.G.); (M.K.S.)
- International Master/Ph.D. Program in Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Research Center of Biomedical Device, Taipei Medical University, Taipei 11031, Taiwan
- Department of Orthopedics, Taipei Medical University–Shuang Ho Hospital, 291 Zhongzheng Rd., Zhonghe District, New Taipei City 11031, Taiwan
| | - Uuganbayar Gankhuyag
- Graduate Institute of Biomedical Materials and Tissue Engineering, School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (B.N.); (Y.B.M.); (U.G.); (M.K.S.)
| | - Andi Pratomo WP
- International Master/Ph.D. Program in Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Mantosh Kumar Satapathy
- Graduate Institute of Biomedical Materials and Tissue Engineering, School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (B.N.); (Y.B.M.); (U.G.); (M.K.S.)
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (B.N.); (Y.B.M.); (U.G.); (M.K.S.)
- Cell Physiology and Molecular Image Research Center, Taipei Medical University–Wan Fang Hospital, 111, Sec. 3, Xinglong 11 Road, Wenshan District, Taipei 116, Taiwan
| |
Collapse
|
222
|
Claeys L, Bravenboer N, Eekhoff EMW, Micha D. Human Fibroblasts as a Model for the Study of Bone Disorders. Front Endocrinol (Lausanne) 2020; 11:394. [PMID: 32636804 PMCID: PMC7318867 DOI: 10.3389/fendo.2020.00394] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 05/18/2020] [Indexed: 01/03/2023] Open
Abstract
Bone tissue degeneration is an urgent clinical issue, making it a subject of intensive research. Chronic skeletal disease forms can be prevalent, such as the age-related osteoporosis, or rare, in the form of monogenetic bone disorders. A barrier in the understanding of the underlying pathological process is the lack of accessibility to relevant material. For this reason, cells of non-bone tissue are emerging as a suitable alternative for models of bone biology. Fibroblasts are highly suitable for this application; they populate accessible anatomical locations, such as the skin tissue. Reports suggesting their utility in preclinical models for the study of skeletal diseases are increasingly becoming available. The majority of these are based on the generation of an intermediate stem cell type, the induced pluripotent stem cells, which are subsequently directed to the osteogenic cell lineage. This intermediate stage is circumvented in transdifferentiation, the process regulating the direct conversion of fibroblasts to osteogenic cells, which is currently not well-explored. With this mini review, we aimed to give an overview of existing osteogenic transdifferentiation models and to inform about their applications in bone biology models.
Collapse
Affiliation(s)
- Lauria Claeys
- Department of Clinical Genetics, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Elisabeth M. W. Eekhoff
- Department of Internal Medicine Section Endocrinology, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Dimitra Micha
- Department of Clinical Genetics, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
223
|
Soluble silica stimulates osteogenic differentiation and gap junction communication in human dental follicle cells. Sci Rep 2020; 10:9923. [PMID: 32555274 PMCID: PMC7303172 DOI: 10.1038/s41598-020-66939-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 05/29/2020] [Indexed: 12/16/2022] Open
Abstract
Several studies have indicated that dietary silicon (Si) is beneficial for bone homeostasis and skeletal health. Furthermore, Si-containing bioactive glass biomaterials have positive effects on bone regeneration when used for repair of bone defects. Si has been demonstrated to stimulate osteoblast differentiation and bone mineralisation in vitro. However, the mechanisms underlying these effects of Si are not well understood. The aim of the present study was to investigate the effects of soluble Si on osteogenic differentiation and connexin 43 (CX43) gap junction communication in cultured pluripotent cells from human dental follicles (hDFC). Neutral Red uptake assay demonstrated that 25 μg/ml of Si significantly stimulated hDFC cell proliferation. Dosages of Si above 100 μg/ml decreased cell proliferation. Alizarin Red staining showed that osteogenic induction medium (OIM) by itself and in combination with Si (25 μg/ml) significantly increased mineralisation in hDFC cultures, although Si alone had no such effect. The expression of osteoblast-related markers in hDFC was analysed with RT-qPCR. OSX, RUNX2, BMP2, ALP, OCN, BSP and CX43 genes were expressed in hDFC cultured for 1, 7, 14 and 21 days. Expression levels of BMP-2 and BSP were significantly upregulated by OIM and Si (25 μg/ml) and were also induced by Si alone. Notably, the expression levels of OCN and CX43 on Day 21 were significantly increased only in the Si group. Flow cytometric measurements revealed that Si (50 μg/ml) significantly increased CX43 protein expression and gap junction communication in hDFC. Next-generation sequencing (NGS) and bioinformatics processing were used for the identification of differentially regulated genes and pathways. The influence of OIM over the cell differentiation profile was more prominent than the influence of Si alone. However, Si in combination with OIM increased the magnitude of expression (up or down) of the differentially regulated genes. The gene for cartilage oligomeric matrix protein (COMP) was the most significantly upregulated. Genes for the regulator of G protein signalling 4 (RGS4), regulator of G protein signalling 2 (RGS2), and matrix metalloproteinases (MMPs) 1, 8, and 10 were also strongly upregulated. Our findings reveal that soluble Si stimulates Cx43 gap junction communication in hDFC and induces gene expression patterns associated with osteogenic differentiation. Taken together, the results support the conclusion that Si is beneficial for bone health.
Collapse
|
224
|
Liu CH, Raj S, Chen CH, Hung KH, Chou CT, Chen IH, Chien JT, Lin IY, Yang SY, Angata T, Tsai WC, Wei JCC, Tzeng IS, Hung SC, Lin KI. HLA-B27-mediated activation of TNAP phosphatase promotes pathogenic syndesmophyte formation in ankylosing spondylitis. J Clin Invest 2020; 129:5357-5373. [PMID: 31682238 DOI: 10.1172/jci125212] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 09/03/2019] [Indexed: 12/31/2022] Open
Abstract
Ankylosing spondylitis (AS) is a type of axial inflammation. Over time, some patients develop spinal ankylosis and permanent disability; however, current treatment strategies cannot arrest syndesmophyte formation completely. Here, we used mesenchymal stem cells (MSCs) from AS patients (AS MSCs) within the enthesis involved in spinal ankylosis to delineate that the HLA-B27-mediated spliced X-box-binding protein 1 (sXBP1)/retinoic acid receptor-β (RARB)/tissue-nonspecific alkaline phosphatase (TNAP) axis accelerated the mineralization of AS MSCs, which was independent of Runt-related transcription factor 2 (Runx2). An animal model mimicking AS pathological bony appositions was established by implantation of AS MSCs into the lumbar spine of NOD-SCID mice. We found that TNAP inhibitors, including levamisole and pamidronate, inhibited AS MSC mineralization in vitro and blocked bony appositions in vivo. Furthermore, we demonstrated that the serum bone-specific TNAP (BAP) level was a potential prognostic biomarker to predict AS patients with a high risk for radiographic progression. Our study highlights the importance of the HLA-B27-mediated activation of the sXBP1/RARB/TNAP axis in AS syndesmophyte pathogenesis and provides a new strategy for the diagnosis and prevention of radiographic progression of AS.
Collapse
Affiliation(s)
- Chin-Hsiu Liu
- Division of Allergy, Immunology and Rheumatology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan.,PhD Program in Translational Medicine, Kaohsiung Medical University and Academia Sinica, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Sengupta Raj
- Royal National Hospital for Rheumatic Diseases, Upper Borough Walls, Bath, United Kingdom
| | - Chun-Hsiung Chen
- Division of Allergy, Immunology and Rheumatology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Kuo-Hsuan Hung
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Chung-Tei Chou
- National Yang-Ming University, Taipei, Taiwan.,Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ing-Ho Chen
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Jui-Teng Chien
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Orthopedics, Chiayi Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi County, Taiwan
| | - I-Ying Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shii-Yi Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Takashi Angata
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Wen-Chan Tsai
- Division of Rheumatology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University and Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - I-Shiang Tzeng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Shih-Chieh Hung
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Integrative Stem Cell Center, Department of Orthopedics, China Medical University Hospital, Taichung, Taiwan.,Institute of New Drug Development, New Drug Development Center, China Medical University, Taichung, Taiwan
| | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
225
|
Lee J, Cha H, Park TH, Park JH. Enhanced osteogenic differentiation of human mesenchymal stem cells by direct delivery of Cbfβ protein. Biotechnol Bioeng 2020; 117:2897-2910. [PMID: 32510167 DOI: 10.1002/bit.27453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/25/2020] [Accepted: 06/04/2020] [Indexed: 12/20/2022]
Abstract
Core binding factor β (Cbfβ) is a non-DNA binding cofactor of Runx2 that potentiates DNA binding. Previously, it has been reported that Cbfβ plays an essential role in osteogenic differentiation and skeletal development by inhibition adipogenesis. Here, we delivered the recombinant Cbfβ protein into human mesenchymal stem cells (MSCs) and triggered osteogenic lineage commitment. The efficient delivery of Cbfβ was achieved by fusing 30Kc19 protein, which is a cell-penetrating protein derived from the silkworm. After the production of the recombinant Cbfβ-30Kc19 protein in the Escherichia coli expression system, and confirmation of its intracellular delivery, MSCs were treated with the Cbfβ-30Kc19 once or twice up to 300 µg/ml. By investigating the upregulation of osteoblast-specific genes and phenotypical changes, such as calcium mineralization, we demonstrated that Cbfβ-30Kc19 efficiently induced osteogenic differentiation in MSCs. At the same time, Cbfβ-30Kc19 suppressed adipocyte formation and downregulated the expression of adipocyte-specific genes. Our results demonstrate that the intracellularly delivered Cbfβ-30Kc19 enhances osteogenesis in MSCs, whereas it suppresses adipogenesis by altering the transcriptional regulatory network involved in osteoblast-adipocyte lineage commitment. Cbfβ-30Kc19 holds great potential for the treatment of bone-related diseases, such as osteoporosis, by allowing transcriptional regulation in MSCs, and overcoming the limitations of current therapies.
Collapse
Affiliation(s)
- Jaeyoung Lee
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Hyeonjin Cha
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Ju Hyun Park
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon-si, Gangwon-do, Republic of Korea
| |
Collapse
|
226
|
Brito Barrera Y, Hause G, Menzel M, Schmelzer C, Lehner E, Mäder K, Wölk C, Groth T. Engineering osteogenic microenvironments by combination of multilayers from collagen type I and chondroitin sulfate with novel cationic liposomes. Mater Today Bio 2020; 7:100071. [PMID: 32924006 PMCID: PMC7476072 DOI: 10.1016/j.mtbio.2020.100071] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 11/28/2022] Open
Abstract
Cationic liposomes composed of a novel lipid (N-{6-amino-1-[N-(9Z) -octadec9-enylamino] -1-oxohexan-(2S) -2-yl} -N'- {2- [N, N-bis(2-aminoethyl) amino] ethyl} -2-hexadecylpropandiamide) (OO4) and dioleoylphosphatidylethanolamine (DOPE) possess high amounts of amino groups and are promising systems for lipofection. Moreover, these cationic liposomes can also be used as a polycationic entity in multilayer formation using layer-by-layer technique (LbL), which is a method to fabricate surface coatings by alternating adsorption of polyanions and polycations. Since liposomes are suitable for endocytosis by or fusion with cells, controlled release of their cargo on site is possible. Here, a polyelectrolyte multilayer (PEM) system was designed of chondroitin sulfate (CS) and collagen type I (Col I) by LbL technique with OO4/DOPE liposomes embedded in the terminal layers to create an osteogenic microenvironment. Both, the composition of PEM and cargo of the liposomes were used to promote osteogenic differentiation of C2C12 myoblasts as in vitro model. The internalization of cargo-loaded liposomes from the PEM into C2C12 cells was studied using lipophilic (Rhodamine-DOPE conjugate) and hydrophilic (Texas Red-labeled dextran) model compounds. Besides, the use of Col I and CS should mimic the extracellular matrix of bone for future applications such as bone replacement therapies. Physicochemical studies of PEM were done to characterize the layer growth, thickness, and topography. The adhesion of myoblast cells was also evaluated whereby the benefit of a cover layer of CS and finally Col I above the liposome layer was demonstrated. As proof of concept, OO4/DOPE liposomes were loaded with dexamethasone, a compound that can induce osteogenic differentiation. A successful induction of osteogenic differentiation of C2C12 cells with the novel designed liposome-loaded PEM system was shown. These findings indicate that designed OH4/DOPE loaded PEMs have a high potential to be used as drug delivery or transfection system for implant coating in the field of bone regeneration and other applications.
Collapse
Key Words
- AFM, Atomic force microscopy
- C2C12 myoblasts
- CLSM, Confocal Laser Scanning Microscopy
- CS, chondroitin sulfate
- Col I, Collagen I
- DLS, Dynamic light scattering
- DMEM, Dulbecco’s modified Eagle’s medium
- DOPE, dioleoylphosphatidylethanolamine
- Dex, Dexamethasone
- ECM, Extracellular matrix
- GAG, Glycosaminoglycan
- LbL, Layer-by-Layer technique
- OO4, (N-{6-amino-1-[N-(9Z) -octadec9-enylamino] -1-oxohexan-(2S) -2-yl} -N’- {2- [N, N-bis(2-aminoethyl) amino] ethyl} -2-hexadecylpropandiamide)
- PBS, Phosphate-buffered saline
- PEI, Polyethylenimine
- PEM, Polyelectrolyte multilayer
- SEM, Scanning electron microscopy
- SPR, Surface plasmon resonance
- TEM, Transmission electron microscopy
- WCA, Water contact angle
- cationic lipids
- chondroitin sulfate
- collagen I
- internalization
- osteogenic differentiation
- polyelectrolyte multilayer system
Collapse
Affiliation(s)
- Y.A. Brito Barrera
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle−Wittenberg, Heinrich Damerow Strasse 4, 06120, Halle (Saale), Germany
| | - G. Hause
- Martin Luther University Halle-Wittenberg, Biocenter, Weinbergweg 22, 06120, Halle (Saale), Germany
| | - M. Menzel
- Department of Biological and Macromolecular Materials, Fraunhofer Institute for Microstructure of Materials and Systems (IMWS), Walter-Hülse-Strasse 1, 06120, Halle (Saale), Germany
| | - C.E.H. Schmelzer
- Department of Biological and Macromolecular Materials, Fraunhofer Institute for Microstructure of Materials and Systems (IMWS), Walter-Hülse-Strasse 1, 06120, Halle (Saale), Germany
| | - E. Lehner
- Department Pharmaceutical Technology, Institute of Pharmacy, Martin Luther University Halle−Wittenberg, Kurt-Mothes Straße 3, 06120, Halle (Saale), Germany
| | - K. Mäder
- Department Pharmaceutical Technology, Institute of Pharmacy, Martin Luther University Halle−Wittenberg, Kurt-Mothes Straße 3, 06120, Halle (Saale), Germany
| | - C. Wölk
- Pharmaceutical Technology, Institute of Pharmacy, Faculty of Medicine, Leipzig University, 04317, Leipzig, Germany
| | - T. 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
| |
Collapse
|
227
|
Avery SJ, Ayre WN, Sloan AJ, Waddington RJ. Interrogating the Osteogenic Potential of Implant SurfacesIn Vitro: A Review of Current Assays. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:217-229. [DOI: 10.1089/ten.teb.2019.0312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Steven James Avery
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Wayne Nishio Ayre
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Alastair James Sloan
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| | - Rachel Jane Waddington
- Department of Oral and Biomedical Sciences, College of Biomedical and Life Sciences, Cardiff Institute for Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
228
|
Sharma AK, Roberts RL, Benson RD, Pierce JL, Yu K, Hamrick MW, McGee-Lawrence ME. The Senolytic Drug Navitoclax (ABT-263) Causes Trabecular Bone Loss and Impaired Osteoprogenitor Function in Aged Mice. Front Cell Dev Biol 2020; 8:354. [PMID: 32509782 PMCID: PMC7252306 DOI: 10.3389/fcell.2020.00354] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/21/2020] [Indexed: 12/02/2022] Open
Abstract
Senescence is a cellular defense mechanism that helps cells prevent acquired damage, but chronic senescence, as in aging, can contribute to the development of age-related tissue dysfunction and disease. Previous studies clearly show that removal of senescent cells can help prevent tissue dysfunction and extend healthspan during aging. Senescence increases with age in the skeletal system, and selective depletion of senescent cells or inhibition of their senescence-associated secretory phenotype (SASP) has been reported to maintain or improve bone mass in aged mice. This suggests that promoting the selective removal of senescent cells, via the use of senolytic agents, can be beneficial in the treatment of aging-related bone loss and osteoporosis. Navitoclax (also known as ABT-263) is a chemotherapeutic drug reported to effectively clear senescent hematopoietic stem cells, muscle stem cells, and mesenchymal stromal cells in previous studies, but its in vivo effects on bone mass had not yet been reported. Therefore, the purpose of this study was to assess the effects of short-term navitoclax treatment on bone mass and osteoprogenitor function in old mice. Aged (24 month old) male and female mice were treated with navitoclax (50 mg/kg body mass daily) for 2 weeks. Surprisingly, despite decreasing senescent cell burden, navitoclax treatment decreased trabecular bone volume fraction in aged female and male mice (−60.1% females, −45.6% males), and BMSC-derived osteoblasts from the navitoclax treated mice were impaired in their ability to produce a mineralized matrix (−88% females, −83% males). Moreover, in vitro administration of navitoclax decreased BMSC colony formation and calcified matrix production by aged BMSC-derived osteoblasts, similar to effects seen with the primary BMSC from the animals treated in vivo. Navitoclax also significantly increased metrics of cytotoxicity in both male and female osteogenic cultures (+1.0 to +11.3 fold). Taken together, these results suggest a potentially harmful effect of navitoclax on skeletal-lineage cells that should be explored further to definitively assess navitoclax’s potential (or risk) as a therapeutic agent for combatting age-related musculoskeletal dysfunction and bone loss.
Collapse
Affiliation(s)
- Anuj K Sharma
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Rachel L Roberts
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Reginald D Benson
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jessica L Pierce
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mark W Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Orthopaedic Surgery, Augusta University, Augusta, GA, United States
| |
Collapse
|
229
|
Rather HA, Patel R, Yadav UCS, Vasita R. Dual drug-delivering polycaprolactone-collagen scaffold to induce early osteogenic differentiation and coupled angiogenesis. ACTA ACUST UNITED AC 2020; 15:045008. [PMID: 32427577 DOI: 10.1088/1748-605x/ab7978] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bone regeneration is a multi-step, overlapping process, in which angiogenesis and osteogenesis are the key players. Several attempts have been made to promote angiogenesis-coupled osteogenesis using scaffolding technology. However, the recreation of functional vasculature during bone regeneration is an unparalleled challenge. In this study, a dual drug-delivering polycaprolactone-collagen fibrous scaffold is reported to promote early osteogenesis and angiogenesis. Simvastatin as a pro-angiogenic and dexamethasone as an osteoinductive drug were encapsulated to functionalize the electrospun fibers. The optically transparent fibrous mat represented the sustained and sequential release of drugs for 28 days. The fibrous mesh increased cell proliferation and enhanced the osteogenic differentiation up to 21 days. The alkaline phosphatase activity and mineral deposition were comparatively higher on dual drug-releasing fibers when compared to control fibers. The dual drug-releasing osteoconductive fibers demonstrated osteogenesis as early as 7 days with a 3.7 and 1.5 fold increase in the expression of osteogenic differentiation markers (RUNX2 and osteocalcin), respectively. In vitro angiogenesis using primary human umbilical vein endothelial cells (pHUVECs) showed no significant difference in cell proliferation among control fibers and dual drug-releasing fibers. However, the angioinductive nature of simvastatin released from the fibers demonstrated tube formation and 2 fold higher angiogenic score. The mRNA and protein expression study of angiogenic markers (VEGFR2 and eNOS) by polymerase chain reaction and western blotting depicted the angioinducing potential of dual drug-releasing fibers. VEGFR2 and eNOS mRNA expressions increased by 1.1 and 1.6 fold, respectively, whereas their protein expression increased by 3.2 and 1.7 fold, respectively. The overall results demonstrate the synergistic effect of osteoconductive substrate and osteoinductive dual drugs to promote early osteogenesis, and release of the pro-angiogenic drug promotes angiogenesis.
Collapse
Affiliation(s)
- Hilal Ahmad Rather
- Biomaterials & Biomimetics laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, 382030 India
| | | | | | | |
Collapse
|
230
|
Li J, Zhao M, Wang Y, Shen M, Wang S, Tang M, Li M, Luo Y, Yang K, Wen X. p75NTR optimizes the osteogenic potential of human periodontal ligament stem cells by up-regulating α1 integrin expression. J Cell Mol Med 2020; 24:7563-7575. [PMID: 32424966 PMCID: PMC7339167 DOI: 10.1111/jcmm.15390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/18/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Human periodontal ligament stem cells (hPDLSCs) are a promising source in regenerative medicine. Due to the complexity and heterogeneity of hPDLSCs, it is critical to isolate homogeneous hPDLSCs with high regenerative potential. In this study, p75 neurotrophin receptor (p75NTR) was used to isolate p75NTR+ and p75NTR− hPDLSCs by fluorescence‐activated cell sorting. Differences in osteogenic differentiation among p75NTR+, p75NTR− and unsorted hPDLSCs were observed. Differential gene expression profiles between p75NTR+ and p75NTR− hPDLSCs were analysed by RNA sequencing. α1 Integrin (ITGA1) small interfering RNA and ITGA1‐overexpressing adenovirus were used to transfect p75NTR+ and p75NTR− hPDLSCs. The results showed that p75NTR+ hPDLSCs demonstrated superior osteogenic capacity than p75NTR− and unsorted hPDLSCs. Differentially expressed genes between p75NTR+ and p75NTR− hPDLSCs were highly involved in the extracellular matrix‐receptor interaction signalling pathway, and p75NTR+ hPDLSCs expressed higher ITGA1 levels than p75NTR− hPDLSCs. ITGA1 silencing inhibited the osteogenic differentiation of p75NTR+ hPDLSCs, while ITGA1 overexpression enhanced the osteogenic differentiation of p75NTR− hPDLSCs. These findings indicate that p75NTR optimizes the osteogenic potential of hPDLSCs by up‐regulating ITGA1 expression, suggesting that p75NTR can be used as a novel cell surface marker to identify and purify hPDLSCs to promote their applications in regenerative medicine.
Collapse
Affiliation(s)
- Jun Li
- Department of Stomatology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Hospital of Stomatology, Zunyi Medical University, Zunyi, China
| | - Manzhu Zhao
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Yingying Wang
- Department of Stomatology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Mengjie Shen
- Hospital of Stomatology, Zunyi Medical University, Zunyi, China
| | - Shuai Wang
- Hospital of Stomatology, Zunyi Medical University, Zunyi, China
| | - Mengying Tang
- Hospital of Stomatology, Southwest Medical University, Luzhou, China
| | - Meng Li
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Yuting Luo
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Kun Yang
- Hospital of Stomatology, Zunyi Medical University, Zunyi, China
| | - Xiujie Wen
- Department of Stomatology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Hospital of Stomatology, Southwest Medical University, Luzhou, China
| |
Collapse
|
231
|
Soltanyzadeh M, Ghollasi M, Halabian R, Shams M. A comparative study of hBM-MSCs' differentiation toward osteogenic lineage in the presence of progesterone and estrogen hormones separately and concurrently in vitro. Cell Biol Int 2020; 44:1701-1713. [PMID: 32339349 DOI: 10.1002/cbin.11364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/12/2020] [Accepted: 04/25/2020] [Indexed: 11/09/2022]
Abstract
Promising cell sources for tissue engineering comprise bone marrow derived-mesenchymal stem cells (BM-MSCs) that have multiple differentiation potentials. Also, sex hormones act as important elements in bone development and maintenance, and the roles of two female sex steroid hormones known as estrogen (17-β estradiol) and progesterone in osteogenic differentiation of human BM-MSCs (hBM-MSCs) are studied. For this purpose, hBM-MSCs were treated with a 1 × 10-6 M concentration of 17-β estradiol and progesterone separately and simultaneously while the optimum concentrations were obtained by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Osteogenic differentiation tests including measurement of alkaline phosphatase (ALP) enzyme activity, the content of total mineral calcium, mineralized matrix staining by Alizarin Red and Von Kossa solutions, real-time reverse transcription polymerase chain reaction (RT-PCR), and immunofluorescence staining were carried out on Days 7 and 14 of differentiation. To exhibit the morphology of the cells, the BM-MSCs were stained with acridine orange (AO) solution. In this study, the results of ALP activity assay, calcium content and real-time RT-PCR assay and also all tests of differentiation staining have shown that 17-β estradiol has been recognized as an enhancing factor of osteogenic differentiation. Furthermore, MTT assay and AO staining revealed progesterone as a factor that seriously improved the proliferation of hBM-MSCs. Generally, the 17-β estradiol individually or in the presence of progesterone has more effects on BM-MSCs' osteogenic differentiation compared to progesterone alone. In this study, it is indicated that the effect of the 17-β estradiol and progesterone concurrently was the same as individual 17-β estradiol on the differentiation of hBM-MSCs.
Collapse
Affiliation(s)
- Maryam Soltanyzadeh
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Marzieh Ghollasi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehdi Shams
- Department of Nanotechnology and Advanced Materials, Materials and Energy Research Center, Karaj, Iran
| |
Collapse
|
232
|
Ramírez Sánchez K, Ledezma-Espinoza A, Sánchez-Kopper A, Avendaño-Soto E, Prado M, Starbird Perez R. Polysaccharide κ-Carrageenan as Doping Agent in Conductive Coatings for Electrochemical Controlled Release of Dexamethasone at Therapeutic Doses. Molecules 2020; 25:molecules25092139. [PMID: 32375224 PMCID: PMC7249122 DOI: 10.3390/molecules25092139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/26/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
Smart conductive materials are developed in regenerative medicine to promote a controlled release profile of charged bioactive agents in the vicinity of implants. The incorporation and the active electrochemical release of the charged compounds into the organic conductive coating is achieved due to its intrinsic electrical properties. The anti-inflammatory drug dexamethasone was added during the polymerization, and its subsequent release at therapeutic doses was reached by electrical stimulation. In this work, a Poly (3,4-ethylenedioxythiophene): κ-carrageenan: dexamethasone film was prepared, and κ-carrageenan was incorporated to keep the electrochemical and physical stability of the electroactive matrix. The presence of κ-carrageenan and dexamethasone in the conductive film was confirmed by µ-Raman spectroscopy and their effect in the topographic was studied using profilometry. The dexamethasone release process was evaluated by cyclic voltammetry and High-Resolution mass spectrometry. In conclusion, κ-carrageenan as a doping agent improves the electrical properties of the conductive layer allowing the release of dexamethasone at therapeutic levels by electrochemical stimulation, providing a stable system to be used in organic bioelectronics systems.
Collapse
Affiliation(s)
- Karla Ramírez Sánchez
- Centro de Investigación y de Servicios Químicos y Microbiológicos (CEQIATEC), School of Chemistry, Instituto Tecnológico de Costa Rica, 159-7050 Cartago, Costa Rica; (A.L.-E.); (A.S.-K.)
- Centro de Investigación en Enfermedades Tropicales (CIET), Faculty of Microbiology, Universidad de Costa Rica, 11501-2060 San José, Costa Rica;
- Correspondence: (K.R.S.); (R.S.P.); Tel.: +506-25502731 (R.S.P.)
| | - Aura Ledezma-Espinoza
- Centro de Investigación y de Servicios Químicos y Microbiológicos (CEQIATEC), School of Chemistry, Instituto Tecnológico de Costa Rica, 159-7050 Cartago, Costa Rica; (A.L.-E.); (A.S.-K.)
| | - Andrés Sánchez-Kopper
- Centro de Investigación y de Servicios Químicos y Microbiológicos (CEQIATEC), School of Chemistry, Instituto Tecnológico de Costa Rica, 159-7050 Cartago, Costa Rica; (A.L.-E.); (A.S.-K.)
| | - Esteban Avendaño-Soto
- Centro de Investigación en Ciencia e Ingeniería de Materiales (CICIMA), Universidad de Costa Rica, 11501-2060 San José, Costa Rica;
- School of Physics, Universidad de Costa Rica, 11501-2060 San José, Costa Rica
| | - Mónica Prado
- Centro de Investigación en Enfermedades Tropicales (CIET), Faculty of Microbiology, Universidad de Costa Rica, 11501-2060 San José, Costa Rica;
| | - Ricardo Starbird Perez
- Centro de Investigación y de Servicios Químicos y Microbiológicos (CEQIATEC), School of Chemistry, Instituto Tecnológico de Costa Rica, 159-7050 Cartago, Costa Rica; (A.L.-E.); (A.S.-K.)
- Correspondence: (K.R.S.); (R.S.P.); Tel.: +506-25502731 (R.S.P.)
| |
Collapse
|
233
|
The voltage-gated proton channel hHv1 is functionally expressed in human chorion-derived mesenchymal stem cells. Sci Rep 2020; 10:7100. [PMID: 32346069 PMCID: PMC7188850 DOI: 10.1038/s41598-020-63517-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/01/2020] [Indexed: 01/08/2023] Open
Abstract
The voltage-gated proton channel Hv1 is widely expressed, among others, in immune and cancer cells, it provides an efficient cytosolic H+extrusion mechanism and regulates vital functions such as oxidative burst, migration and proliferation. Here we demonstrate the presence of human Hv1 (hHv1) in the placenta/chorion-derived mesenchymal stem cells (cMSCs) using RT-PCR. The voltage- and pH-dependent gating of the current is similar to that of hHv1 expressed in cell lines and that the current is blocked by 5-chloro-2-guanidinobenzimidazole (ClGBI) and activated by arachidonic acid (AA). Inhibition of hHv1 by ClGBI significantly decreases mineral matrix production of cMSCs induced by conditions mimicking physiological or pathological (inorganic phosphate, Pi) induction of osteogenesis. Wound healing assay and single cell motility analysis show that ClGBI significantly inhibits the migration of cMSCs. Thus, seminal functions of cMSCs are modulated by hHv1 which makes this channel as an attractive target for controlling advantages/disadvantages of MSCs therapy.
Collapse
|
234
|
Nasello G, Alamán-Díez P, Schiavi J, Pérez MÁ, McNamara L, García-Aznar JM. Primary Human Osteoblasts Cultured in a 3D Microenvironment Create a Unique Representative Model of Their Differentiation Into Osteocytes. Front Bioeng Biotechnol 2020; 8:336. [PMID: 32391343 PMCID: PMC7193048 DOI: 10.3389/fbioe.2020.00336] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/26/2020] [Indexed: 01/12/2023] Open
Abstract
Microengineered systems provide an in vitro strategy to explore the variability of individual patient response to tissue engineering products, since they prefer the use of primary cell sources representing the phenotype variability. Traditional in vitro systems already showed that primary human osteoblasts embedded in a 3D fibrous collagen matrix differentiate into osteocytes under specific conditions. Here, we hypothesized that translating this environment to the organ-on-a-chip scale creates a minimal functional unit to recapitulate osteoblast maturation toward osteocytes and matrix mineralization. Primary human osteoblasts were seeded in a type I collagen hydrogel, to establish the role of lower (2.5 × 105 cells/ml) and higher (1 × 106 cells/ml) cell density on their differentiation into osteocytes. A custom semi-automatic image analysis software was used to extract quantitative data on cellular morphology from brightfield images. The results are showing that cells cultured at a high density increase dendrite length over time, stop proliferating, exhibit dendritic morphology, upregulate alkaline phosphatase (ALP) activity, and express the osteocyte marker dental matrix protein 1 (DMP1). On the contrary, cells cultured at lower density proliferate over time, do not upregulate ALP and express the osteoblast marker bone sialoprotein 2 (BSP2) at all timepoints. Our work reveals that microengineered systems create unique conditions to capture the major aspects of osteoblast differentiation into osteocytes with a limited number of cells. We propose that the microengineered approach is a functional strategy to create a patient-specific bone tissue model and investigate the individual osteogenic potential of the patient bone cells.
Collapse
Affiliation(s)
- Gabriele Nasello
- Multiscale in Mechanical and Biological Engineering (M2BE), University of Zaragoza, Zaragoza, Spain.,Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Pilar Alamán-Díez
- Multiscale in Mechanical and Biological Engineering (M2BE), University of Zaragoza, Zaragoza, Spain
| | - Jessica Schiavi
- Mechanobiology and Medical Device Research Group (MMDRG), National University of Ireland Galway, Galway, Ireland
| | - María Ángeles Pérez
- Multiscale in Mechanical and Biological Engineering (M2BE), University of Zaragoza, Zaragoza, Spain
| | - Laoise McNamara
- Mechanobiology and Medical Device Research Group (MMDRG), National University of Ireland Galway, Galway, Ireland
| | - José Manuel García-Aznar
- Multiscale in Mechanical and Biological Engineering (M2BE), University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
235
|
Zhang XT, Sun M, Zhang L, Dai YK, Wang F. The potential function of miR-135b-mediated JAK2/STAT3 signaling pathway during osteoblast differentiation. Kaohsiung J Med Sci 2020; 36:673-681. [PMID: 32319222 DOI: 10.1002/kjm2.12217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/31/2020] [Accepted: 03/23/2020] [Indexed: 11/09/2022] Open
Abstract
MC3T3-E1 cells were divided into Blank, miR-135b mimics, miR-135b inhibitors, AG490, and miR-135b inhibitors + AG490 groups. Cell viability was determined by MTT, alkaline phosphatase (ALP) activity by the corresponding kit, and mineralization by alizarin red staining. Furthermore, miR-135b, osteoblast-specific genes, and JAK2/STAT3 were detected through quantitative real-time polymerase chain reaction and Western blotting. MiR-135b downregulation was identified with increased JAK2 during osteoblast differentiation. JAK2 was confirmed as a target gene of miR-135b by dual-luciferase reporter assay. MC3T3-E1 cells in both miR-135b mimics and AG490 groups manifested decrease in cell viability, ALP activity, and mineralized nodes, as well as reductions in osteoblast-specific genes and proteins of JAK2, p-JAK2, and p-STAT3, but increase in cell apoptosis. However, opposite changes of the above factors were shown in cells from miR-135b inhibitors group. Notably, AG490 could reverse promotion effects of miR-135b inhibitors on osteoblast differentiation. Inhibiting miR-135b could activate the JAK2/STAT3 signaling pathway, thereby improving the cell viability and promoting the osteoblast differentiation.
Collapse
Affiliation(s)
- Xiang-Tao Zhang
- Department of Orthopedics, The No.1 Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Min Sun
- Department of Neonatology, Children's Hospital of Hebei Province, Shijiazhuang, Hebei, China
| | - Li Zhang
- The second Department of Orthopedics, The Third Hospital of Hebei Medical University & You Yi Branch, Shijiazhuang, Hebei, China
| | - Yi-Ke Dai
- The first Department of Arthrosis, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fei Wang
- The first Department of Arthrosis, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
236
|
Shafaei H, Kalarestaghi H. Adipose-derived stem cells: An appropriate selection for osteogenic differentiation. J Cell Physiol 2020; 235:8371-8386. [PMID: 32239731 DOI: 10.1002/jcp.29681] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are a major component of various forms of tissue engineering. MSCs have self-renewal and multidifferential potential. Osteogenic differentiation of MSCs is an area of attention in bone regeneration. One form of MSCs are adipose-derived stem cells (ASCs), which can be simply harvested and differentiated into several cell lineages, such as chondrocytes, adipocytes, or osteoblasts. Due to special properties, ASCs are frequently used in vitro and in vivo bone regeneration. Identifying factors involved in osteogenic differentiation of ASCs is important for better understanding the mechanism of osteogenic differentiation. Different methods are used to stimulate osteogenesis of ASCs in literature, including common osteogenic media, growth factors, hormones, hypoxia, mechanical and chemical stimuli, genetic modification, and nanotechnology. This review article provides an overview describing the isolation procedure, characterization, properties, current methods for osteogenic differentiation of ASCs, and their basic biological mechanism.
Collapse
Affiliation(s)
- Hajar Shafaei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Kalarestaghi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran.,Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences and Pathology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
237
|
Di Carlo R, Di Crescenzo A, Pilato S, Ventrella A, Piattelli A, Recinella L, Chiavaroli A, Giordani S, Baldrighi M, Camisasca A, Zavan B, Falconi M, Cataldi A, Fontana A, Zara S. Osteoblastic Differentiation on Graphene Oxide-Functionalized Titanium Surfaces: An In Vitro Study. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E654. [PMID: 32244572 PMCID: PMC7221922 DOI: 10.3390/nano10040654] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/20/2020] [Accepted: 03/26/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND Titanium implant surfaces are continuously modified to improve biocompatibility and to promote osteointegration. Graphene oxide (GO) has been successfully used to ameliorate biomaterial performances, in terms of implant integration with host tissue. The aim of this study is to evaluate the Dental Pulp Stem Cells (DPSCs) viability, cytotoxic response, and osteogenic differentiation capability in the presence of GO-coated titanium surfaces. METHODS Two titanium discs types, machined (control, Crtl) and sandblasted and acid-etched (test, Test) discs, were covalently functionalized with GO. The ability of the GO-functionalized substrates to allow the proliferation and differentiation of DPSCs, as well as their cytotoxic potential, were assessed. RESULTS The functionalization procedures provide a homogeneous coating with GO of the titanium surface in both control and test substrates, with unchanged surface roughness with respect to the untreated surfaces. All samples show the deposition of extracellular matrix, more pronounced in the test and GO-functionalized test discs. GO-functionalized test samples evidenced a significant viability, with no cytotoxic response and a remarkable early stage proliferation of DPSCs cells, followed by their successful differentiation into osteoblasts. CONCLUSIONS The described protocol of GO-functionalization provides a novel not cytotoxic biomaterial that is able to stimulate cell viability and that better and more quickly induces osteogenic differentiation with respect to simple titanium discs. Our findings pave the way to exploit this GO-functionalization protocol for the production of novel dental implant materials that display improved integration with the host tissue.
Collapse
Affiliation(s)
- Roberta Di Carlo
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy
| | - Antonello Di Crescenzo
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy
| | - Serena Pilato
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy
| | - Alessia Ventrella
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy
| | - Adriano Piattelli
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy
| | - Lucia Recinella
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy
| | - Annalisa Chiavaroli
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy
| | - Silvia Giordani
- School of Chemical Sciences, Dublin City University, Glasnevin, D09 E432 Dublin 9, Ireland
| | - Michele Baldrighi
- Nano Carbon Materials, Italian Institute of Technology, via Morego 30, 16163 Genova, Italy
| | - Adalberto Camisasca
- School of Chemical Sciences, Dublin City University, Glasnevin, D09 E432 Dublin 9, Ireland
| | - Barbara Zavan
- Medical Science Department, University of Ferrara, via Aldo Moro 8, 44121 Ferrara, Italy
| | - Mirella Falconi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Amelia Cataldi
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy
| | - Antonella Fontana
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy
| | - Susi Zara
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
238
|
Hung BP, Gonzalez-Fernandez T, Lin JB, Campbell T, Lee YB, Panitch A, Alsberg E, Leach JK. Multi-peptide presentation and hydrogel mechanics jointly enhance therapeutic duo-potential of entrapped stromal cells. Biomaterials 2020; 245:119973. [PMID: 32244091 DOI: 10.1016/j.biomaterials.2020.119973] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/27/2022]
Abstract
The native extracellular matrix (ECM) contains a host of matricellular proteins and bioactive factors that regulate cell behavior, and many ECM components have been leveraged to guide cell fate. However, the large size and chemical characteristics of these constituents complicate their incorporation into biomaterials without interfering with material properties, motivating the need for alternative approaches to regulate cellular responses. Mesenchymal stromal cells (MSCs) can promote osseous regeneration in vivo directly or indirectly through multiple means including (1) secretion of proangiogenic and mitogenic factors to initiate formation of a vascular template and recruit host cells into the tissue site or (2) direct differentiation into osteoblasts. As MSC behavior is influenced by the properties of engineered hydrogels, we hypothesized that the biochemical and biophysical properties of alginate could be manipulated to promote the dual contributions of encapsulated MSCs toward bone formation. We functionalized alginate with QK peptide to enhance proangiogenic factor secretion and RGD to promote adhesion, while biomechanical-mediated osteogenic cues were controlled by modulating viscoelastic properties of the alginate substrate. A 1:1 ratio of QK:RGD resulted in the highest levels of both proangiogenic factor secretion and mineralization in vitro. Viscoelastic alginate outperformed purely elastic gels in both categories, and this effect was enhanced by stiffness up to 20 kPa. Furthermore, viscoelastic constructs promoted vessel infiltration and bone regeneration in a rat calvarial defect over 12 weeks. These data suggest that modulating viscoelastic properties of biomaterials, in conjunction with dual peptide functionalization, can simultaneously enhance multiple aspects of MSC regenerative potential and improve neovascularization of engineered tissues.
Collapse
Affiliation(s)
- Ben P Hung
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | | | - Jenny B Lin
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Takeyah Campbell
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Yu Bin Lee
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, USA
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, USA
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA; Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, USA.
| |
Collapse
|
239
|
Tetè G, Capparè P, Gherlone E. New Application of Osteogenic Differentiation from HiPS Stem Cells for Evaluating the Osteogenic Potential of Nanomaterials in Dentistry. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17061947. [PMID: 32188154 PMCID: PMC7142891 DOI: 10.3390/ijerph17061947] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/14/2022]
Abstract
Objective: HiPS stem cells are commonly used for the study of medical disorders. The laboratory in which this study was conducted uses these cells for examining the treatment and cure of neurodegenerative diseases. Bone regeneration poses the greatest challenge for an oral surgeon both in terms of increased implant osseointegration and reducing bone healing times. The aim of this study was to validate the protocol in the literature to produce and then test in vitro osteoblasts with different nanomaterials to simulate bone regeneration. Method: hiPS clones (#2, #4, and #8) were differentiated into an osteoblast cell culture tested for alizarin red staining and for alkaline phosphatase testing at 14, 21 and 28 days, after the cells were plated. Results: The cells showed diffuse positivity under alizarin red staining and the alkaline phosphatase (ALP)-test, showing small formations of calcium clusters. Conclusion: Despite the limitations of our study, it is a starting point for further protocols, laying a solid foundation for research in the field of bone regeneration through the use of stem cells.
Collapse
Affiliation(s)
- Giulia Tetè
- Specialization School in Oral Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Correspondence: (G.T.); (P.C.); Tel.: +39-02-2643-3022 (G.T.)
| | - Paolo Capparè
- Dental School, Vita-Salute University and IRCCS San Raffaele, 20132 Milan, Italy;
- Correspondence: (G.T.); (P.C.); Tel.: +39-02-2643-3022 (G.T.)
| | - Enrico Gherlone
- Dental School, Vita-Salute University and IRCCS San Raffaele, 20132 Milan, Italy;
| |
Collapse
|
240
|
Uribe-Etxebarria V, García-Gallastegui P, Pérez-Garrastachu M, Casado-Andrés M, Irastorza I, Unda F, Ibarretxe G, Subirán N. Wnt-3a Induces Epigenetic Remodeling in Human Dental Pulp Stem Cells. Cells 2020; 9:cells9030652. [PMID: 32156036 PMCID: PMC7140622 DOI: 10.3390/cells9030652] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022] Open
Abstract
Dental pulp stem cells (DPSCs) from adult teeth show the expression of a very complete repertoire of stem pluripotency core factors and a high plasticity for cell reprogramming. Canonical Wnt and Notch signaling pathways regulate stemness and the expression of pluripotency core factors in DPSCs, and even very short-term (48 h) activations of the Wnt pathway induce a profound remodeling of DPSCs at the physiologic and metabolic levels. In this work, DPSC cultures were exposed to treatments modulating Notch and Wnt signaling, and also induced to differentiate to osteo/adipocytes. DNA methylation, histone acetylation, histone methylation, and core factor expression levels where assessed by mass spectroscopy, Western blot, and qPCR. A short-term activation of Wnt signaling by WNT-3A induced a genomic DNA demethylation, and increased histone acetylation and histone methylation in DPSCs. The efficiency of cell reprogramming methods relies on the ability to surpass the epigenetic barrier, which determines cell lineage specificity. This study brings important information about the regulation of the epigenetic barrier by Wnt signaling in DPSCs, which could contribute to the development of safer and less aggressive reprogramming methodologies with a view to cell therapy.
Collapse
Affiliation(s)
- Verónica Uribe-Etxebarria
- Cell Biology and Histology Department, University of the Basque Country (UPV/EHU), Barrio Sarriena, S/N, 48940 Leioa, Spain; (V.U.-E.); (P.G.-G.); (M.P.-G.); (M.C.-A.); (I.I.); (F.U.)
- Pathology Department, New York University, 550 1st Avenue, New York, NY 10016, USA
| | - Patricia García-Gallastegui
- Cell Biology and Histology Department, University of the Basque Country (UPV/EHU), Barrio Sarriena, S/N, 48940 Leioa, Spain; (V.U.-E.); (P.G.-G.); (M.P.-G.); (M.C.-A.); (I.I.); (F.U.)
| | - Miguel Pérez-Garrastachu
- Cell Biology and Histology Department, University of the Basque Country (UPV/EHU), Barrio Sarriena, S/N, 48940 Leioa, Spain; (V.U.-E.); (P.G.-G.); (M.P.-G.); (M.C.-A.); (I.I.); (F.U.)
| | - María Casado-Andrés
- Cell Biology and Histology Department, University of the Basque Country (UPV/EHU), Barrio Sarriena, S/N, 48940 Leioa, Spain; (V.U.-E.); (P.G.-G.); (M.P.-G.); (M.C.-A.); (I.I.); (F.U.)
- Unité Mixte de Recherche UMR1029. INSERM-Université de Bordeaux, 33000 Bordeaux, France
| | - Igor Irastorza
- Cell Biology and Histology Department, University of the Basque Country (UPV/EHU), Barrio Sarriena, S/N, 48940 Leioa, Spain; (V.U.-E.); (P.G.-G.); (M.P.-G.); (M.C.-A.); (I.I.); (F.U.)
| | - Fernando Unda
- Cell Biology and Histology Department, University of the Basque Country (UPV/EHU), Barrio Sarriena, S/N, 48940 Leioa, Spain; (V.U.-E.); (P.G.-G.); (M.P.-G.); (M.C.-A.); (I.I.); (F.U.)
| | - Gaskon Ibarretxe
- Cell Biology and Histology Department, University of the Basque Country (UPV/EHU), Barrio Sarriena, S/N, 48940 Leioa, Spain; (V.U.-E.); (P.G.-G.); (M.P.-G.); (M.C.-A.); (I.I.); (F.U.)
- Correspondence: ; Tel.: +34-94-601-3218
| | - Nerea Subirán
- Physiology Department, University of the Basque Country (UPV/EHU), Barrio Sarriena, S/N, 48940 Leioa, Spain;
| |
Collapse
|
241
|
Kuterbekov M, Jonas AM, Glinel K, Picart C. Osteogenic Differentiation of Adipose-Derived Stromal Cells: From Bench to Clinics. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:461-474. [PMID: 32098603 DOI: 10.1089/ten.teb.2019.0225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In addition to mesenchymal stem cells, adipose-derived stem/stromal cells (ASCs) are an attractive source for a large variety of cell-based therapies. One of their most important potential applications is related to the regeneration of bone tissue thanks to their capacity to differentiate in bone cells. However, this requires a proper control of their osteogenic differentiation, which depends not only on the initial characteristics of harvested cells but also on the conditions used for their culture. In this review, we first briefly describe the preclinical and clinical trials using ASCs for bone regeneration and present the quantitative parameters used to characterize the osteogenic differentiation of ASCs. We then focus on the soluble factors influencing the osteogenic differentiation of ACS, including the steroid hormones and various growth factors, notably the most osteoinductive ones, the bone morphogenetic proteins (BMPs). Impact statement Adipose-derived stromal/stem cells are reviewed for their use in bone regeneration.
Collapse
Affiliation(s)
- Mirasbek Kuterbekov
- Institute of Condensed Matter & Nanosciences (Bio & Soft Matter), Université Catholique de Louvain, Louvain-la-Neuve, Belgium.,Grenoble Institute of Technology, University Grenoble Alpes, LMGP, Grenoble, France
| | - Alain M Jonas
- Institute of Condensed Matter & Nanosciences (Bio & Soft Matter), Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Karine Glinel
- Institute of Condensed Matter & Nanosciences (Bio & Soft Matter), Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Catherine Picart
- Grenoble Institute of Technology, University Grenoble Alpes, LMGP, Grenoble, France.,Biomimetism and Regenerative Medicine Lab, CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Université Grenoble-Alpes/CEA/CNRS, Grenoble, France
| |
Collapse
|
242
|
Owen R, Bahmaee H, Claeyssens F, Reilly GC. Comparison of the Anabolic Effects of Reported Osteogenic Compounds on Human Mesenchymal Progenitor-derived Osteoblasts. Bioengineering (Basel) 2020; 7:E12. [PMID: 31972962 PMCID: PMC7148480 DOI: 10.3390/bioengineering7010012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 01/10/2023] Open
Abstract
There is variability in the reported effects of compounds on osteoblasts arising from differences in experimental design and choice of cell type/origin. This makes it difficult to discern a compound's action outside its original study and compare efficacy between compounds. Here, we investigated five compounds frequently reported as anabolic for osteoblasts (17β-estradiol (oestrogen), icariin, lactoferrin, lithium chloride, and menaquinone-4 (MK-4)) on human mesenchymal progenitors to assess their potential for bone tissue engineering with the aim of identifying a potential alternative to expensive recombinant growth factors such as bone morphogenetic protein 2 (BMP-2). Experiments were performed using the same culture conditions to allow direct comparison. The concentrations of compounds spanned two orders of magnitude to encompass the reported efficacious range and were applied continuously for 22 days. The effects on the proliferation (resazurin reduction and DNA quantification), osteogenic differentiation (alkaline phosphatase (ALP) activity), and mineralised matrix deposition (calcium and collagen quantification) were assessed. Of these compounds, only 10 µM MK-4 stimulated a significant anabolic response with 50% greater calcium deposition. Oestrogen and icariin had no significant effects, with the exception of 1 µM icariin, which increased the metabolic activity on days 8 and 22. 1000 µg/mL of lactoferrin and 10 mM lithium chloride both significantly reduced the mineralised matrix deposition in comparison to the vehicle control, despite the ALP activity being higher in lithium chloride-treated cells at day 15. This demonstrates that MK-4 is the most powerful stimulant of bone formation in hES-MPs of the compounds investigated, highlighting its potential in bone tissue engineering as a method of promoting bone formation, as well as its prospective use as an osteoporosis treatment.
Collapse
Affiliation(s)
- Robert Owen
- Department of Materials Science and Engineering, INSIGNEO Institute for In Silico Medicine, The Pam Liversidge Building, Sir Frederick Mappin Building, Mappin Street, Sheffield S1 3JD, UK; (H.B.); (F.C.); (G.C.R.)
- Department of Materials Science and Engineering, University of Sheffield, Kroto Research Institute, Sheffield S3 7HQ, UK
| | - Hossein Bahmaee
- Department of Materials Science and Engineering, INSIGNEO Institute for In Silico Medicine, The Pam Liversidge Building, Sir Frederick Mappin Building, Mappin Street, Sheffield S1 3JD, UK; (H.B.); (F.C.); (G.C.R.)
- Department of Materials Science and Engineering, University of Sheffield, Kroto Research Institute, Sheffield S3 7HQ, UK
| | - Frederik Claeyssens
- Department of Materials Science and Engineering, INSIGNEO Institute for In Silico Medicine, The Pam Liversidge Building, Sir Frederick Mappin Building, Mappin Street, Sheffield S1 3JD, UK; (H.B.); (F.C.); (G.C.R.)
- Department of Materials Science and Engineering, University of Sheffield, Kroto Research Institute, Sheffield S3 7HQ, UK
| | - Gwendolen C. Reilly
- Department of Materials Science and Engineering, INSIGNEO Institute for In Silico Medicine, The Pam Liversidge Building, Sir Frederick Mappin Building, Mappin Street, Sheffield S1 3JD, UK; (H.B.); (F.C.); (G.C.R.)
| |
Collapse
|
243
|
Alksne M, Kalvaityte M, Simoliunas E, Rinkunaite I, Gendviliene I, Locs J, Rutkunas V, Bukelskiene V. In vitro comparison of 3D printed polylactic acid/hydroxyapatite and polylactic acid/bioglass composite scaffolds: Insights into materials for bone regeneration. J Mech Behav Biomed Mater 2020; 104:103641. [PMID: 32174399 DOI: 10.1016/j.jmbbm.2020.103641] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/13/2019] [Accepted: 01/14/2020] [Indexed: 02/07/2023]
Abstract
3D printing of polylactic acid (PLA) and hydroxyapatite (HA) or bioglass (BG) bioceramics composites is the most promising technique for artificial bone construction. However, HA and BG have different chemical composition as well as different bone regeneration inducing mechanisms. Thus, it is important to compare differentiation processes induced by 3D printed PLA + HA and PLA + BG scaffolds in order to evaluate the strongest osteoconductive and osteoinductive properties possessing bioceramics. In this study, we analysed porous PLA + HA (10%) and PLA + BG (10%) composites' effect on rat's dental pulp stem cells fate in vitro. Obtained results indicated, that PLA + BG scaffolds lead to weaker cell adhesion and proliferation than PLA + HA. Nevertheless, osteoinductive and other biofriendly properties were more pronounced by PLA + BG composites. Overall, the results showed a strong advantage of bioceramic BG against HA, thus, 3D printed PLA + BG composite scaffolds could be a perspective component for patient-specific, cheaper and faster artificial bone tissue production.
Collapse
Affiliation(s)
- Milda Alksne
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257, Vilnius, Lithuania.
| | - Migle Kalvaityte
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257, Vilnius, Lithuania
| | - Egidijus Simoliunas
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257, Vilnius, Lithuania
| | - Ieva Rinkunaite
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257, Vilnius, Lithuania
| | - Ieva Gendviliene
- Institute of Odontology, Faculty of Medicine, Vilnius University, Zalgirio Str. 115, LT-08217, Vilnius, Lithuania
| | - Janis Locs
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka 3, Riga, LV-1007, Latvia
| | - Vygandas Rutkunas
- Institute of Odontology, Faculty of Medicine, Vilnius University, Zalgirio Str. 115, LT-08217, Vilnius, Lithuania
| | - Virginija Bukelskiene
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257, Vilnius, Lithuania
| |
Collapse
|
244
|
Leite DM, Sousa DM, Lamghari M, Pêgo AP. Exploring Poly(Ethylene Glycol)-Poly(Trimethylene Carbonate) Nanoparticles as Carriers of Hydrophobic Drugs to Modulate Osteoblastic Activity. J Pharm Sci 2020; 109:1594-1604. [PMID: 31935391 DOI: 10.1016/j.xphs.2020.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/07/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022]
Abstract
Current treatment options for bone-related disorders rely on a systemic administration of therapeutic agents that possess low solubility and intracellular bioavailability, as well as a high pharmacokinetic variability, which in turn lead to major off-target side effects. Hence, there is an unmet need of developing drug delivery systems that can improve the clinical efficacy of such therapeutic agents. Nanoparticle delivery systems might serve as promising carriers of hydrophobic molecules. Here, we propose 2 nanoparticle-based delivery systems based on monomethoxy poly(ethylene glycol)-poly(trimethyl carbonate) (mPEG-PTMC) and poly(lactide-co-glycolide) for the intracellular controlled release of a small hydrophobic drug (dexamethasone) to osteoblast cells in vitro. mPEG-PTMC self-assembles into stable nanoparticles in the absence of surfactant and shows a greater entrapment capacity of dexamethasone, while assuring bioactivity in MC3T3-E1 and bone marrow stromal cells cultured under apoptotic and osteogenic conditions, respectively. The mPEG-PTMC nanoparticles represent a potential vector for the intracellular delivery of hydrophobic drugs in the framework of bone-related diseases.
Collapse
Affiliation(s)
- Diana M Leite
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, R. Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Daniela M Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Meriem Lamghari
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, R. de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Ana Paula Pêgo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, R. Dr. Roberto Frias s/n, 4200-465 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, R. de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
245
|
Suryani L, Too JH, Hassanbhai AM, Wen F, Lin DJ, Yu N, Teoh SH. Effects of Electromagnetic Field on Proliferation, Differentiation, and Mineralization of MC3T3 Cells. Tissue Eng Part C Methods 2020; 25:114-125. [PMID: 30661463 DOI: 10.1089/ten.tec.2018.0364] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
IMPACT STATEMENT We present the study about how the parameters of pulsed electromagnetic field (PEMF) stimulus affected calvarial osteoblast precursor cell in terms of growth, viability, and differentiation. This research provides insight and foundation to clinical application of noninvasive therapy using PEMF to improve bone regeneration.
Collapse
Affiliation(s)
- Luvita Suryani
- 1 School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.,2 Centre for Developmental Biology, Tissue Engineering, Regenerative Medicine and Innovation, Singapore, Singapore
| | - Jian Hui Too
- 3 National Dental Centre Singapore, Singapore, Singapore
| | - Ammar Mansoor Hassanbhai
- 1 School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.,2 Centre for Developmental Biology, Tissue Engineering, Regenerative Medicine and Innovation, Singapore, Singapore
| | - Feng Wen
- 1 School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.,2 Centre for Developmental Biology, Tissue Engineering, Regenerative Medicine and Innovation, Singapore, Singapore
| | - Daryl Jimian Lin
- 1 School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Na Yu
- 3 National Dental Centre Singapore, Singapore, Singapore.,4 Duke-NUS Medical School Singapore, Singapore, Singapore
| | - Swee-Hin Teoh
- 1 School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.,2 Centre for Developmental Biology, Tissue Engineering, Regenerative Medicine and Innovation, Singapore, Singapore.,5 Lee Kong Chian School of Medicine Singapore, Singapore, Singapore
| |
Collapse
|
246
|
Sunarso, Tsuchiya A, Toita R, Tsuru K, Ishikawa K. Enhanced Osseointegration Capability of Poly(ether ether ketone) via Combined Phosphate and Calcium Surface-Functionalization. Int J Mol Sci 2019; 21:E198. [PMID: 31892154 PMCID: PMC6981423 DOI: 10.3390/ijms21010198] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/24/2019] [Accepted: 12/25/2019] [Indexed: 02/06/2023] Open
Abstract
Biomedical applications of poly(ether ether ketone) (PEEK) are hindered by its inherent bioinertness and lack of osseointegration capability. In the present study, to enhance osteogenic activity and, hence, the osseointegration capability of PEEK, we proposed a strategy of combined phosphate and calcium surface-functionalization, in which ozone-gas treatment and wet chemistry were used for introduction of hydroxyl groups and modification of phosphate and/or calcium, respectively. Surface functionalization significantly elevated the surface hydrophilicity without changing the surface roughness or topography. The cell study demonstrated that immobilization of phosphate or calcium increased the osteogenesis of rat mesenchymal stem cells compared with bare PEEK, including cell proliferation, alkaline phosphatase activity, and bone-like nodule formation. Interestingly, further enhancement was observed for samples co-immobilized with phosphate and calcium. Furthermore, in the animal study, phosphate and calcium co-functionalized PEEK demonstrated significantly enhanced osseointegration, as revealed by a greater direct bone-to-implant contact ratio and bond strength between the bone and implant than unfunctionalized and phosphate-functionalized PEEK, which paves the way for the orthopedic and dental application of PEEK.
Collapse
Affiliation(s)
- Sunarso
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (S.); (A.T.); (K.T.); (K.I.)
- Department of Dental Materials, Faculty of Dentistry, Universitas Indonesia, Jalan Salemba Raya No. 4, Jakarta 10430, Indonesia
| | - Akira Tsuchiya
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (S.); (A.T.); (K.T.); (K.I.)
| | - Riki Toita
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (S.); (A.T.); (K.T.); (K.I.)
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan
| | - Kanji Tsuru
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (S.); (A.T.); (K.T.); (K.I.)
- Section of Bioengineering, Department of Dental Engineering, Fukuoka Dental College, 2-15-1 Tamura, Sawara, Fukuoka 814-0193, Japan
| | - Kunio Ishikawa
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (S.); (A.T.); (K.T.); (K.I.)
| |
Collapse
|
247
|
Leslie SK, Cohen DJ, Boyan BD, Schwartz Z. Production of osteogenic and angiogenic factors by microencapsulated adipose stem cells varies with culture conditions. J Biomed Mater Res B Appl Biomater 2019; 108:1857-1867. [PMID: 31872938 DOI: 10.1002/jbm.b.34527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/28/2019] [Accepted: 11/16/2019] [Indexed: 01/06/2023]
Abstract
Growth factors produced by stem cells aid in the bone repair process. We investigated the ability of encapsulated rat adipose-derived stem cells (rASCs) treated with osteogenic media (OM) to produce growth factors, and determined the optimal combination of OM components that will lead to the production of both osteogenic and angiogenic factors. Our results demonstrate that microencapsulated stem cells were able to produce vascular endothelial growth factor (VEGF), fibroblast growth factor-2, and bone morphogenetic protein-2 (BMP2) necessary for bone regeneration. OM led to the reduction of angiogenic factors; however, the removal of dexamethasone restored angiogenic factor production. Additionally, we determined whether the effect of dexamethasone on VEGF and BMP2 varied among rat, rabbit, mouse, and humans. Dexamethasone led to a reduction in VEGF levels in ASCs derived from rats, mice, and humans, while this reduction was absent in rabbit ASCs (rbASCs). Human ASCs (hASCs) from donors of different race and sex showed a similar response to dexamethasone with secreted VEGF levels. BMP2 levels secreted by rbASCs, mouse ASCs (mASCs), and hASCs were independent of the media treatments, while rASCs responded differently in the surrounding media and within the microbeads. In conclusion, microencapsulated ASCs can be treated to produce osteogenic and angiogenic factors for tissue regeneration applications, but outcomes may vary with culture conditions.
Collapse
Affiliation(s)
- Shirae K Leslie
- College of Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - David Joshua Cohen
- College of Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Barbara D Boyan
- College of Engineering, Virginia Commonwealth University, Richmond, Virginia.,Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, Atlanta, Georgia
| | - Zvi Schwartz
- College of Engineering, Virginia Commonwealth University, Richmond, Virginia.,Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
248
|
Evaluation of Chitosan Hydrogel for Sustained Delivery of VEGF for Odontogenic Differentiation of Dental Pulp Stem Cells. Stem Cells Int 2019; 2019:1515040. [PMID: 31949434 PMCID: PMC6942904 DOI: 10.1155/2019/1515040] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/14/2019] [Accepted: 11/25/2019] [Indexed: 02/05/2023] Open
Abstract
The pulpotomy with pulp capping is aimed at retaining vital pulp with reparative dentin formation. Vascular endothelial growth factor (VEGF) plays a crucial role in dentin regeneration; however, its constant administrations in the human body is still problematic. Chitosan was widely studied as an effective carrier to deliver bioactive molecules in regenerative medicine. In this study, we conducted a chitosan/β-glycerophosphate (CS/β-GP) hydrogel as a VEGF-sustained release system and explored its effects on dental pulp stem cells (DPSCs). CS/β-GP hydrogel was manufactured using a sol-gel method. SEM assay showed the spongy and porous microstructure of the lyophilized hydrogels. DPSCs cultured in the CS/β-GP hydrogel kept adhesion and vitality. CCK-8 assay tested the promoted proliferation activity of DPSCs on the hydrogel. Besides, the added VEGF protein could continually release from VEGF/CS/β-GP hydrogel. The VEGF/CS/β-GP hydrogel could promote the odontogenic differentiation of DPSCs better than VEGF treatment without hydrogel. Our results suggested that CS/β-GP hydrogel could continually release VEGF and contribute to odontogenic differentiation of DPSCs, thus may become a potential carrier of bioactive molecules in pulp capping therapy.
Collapse
|
249
|
Induction of aortic valve calcification by celecoxib and its COX-2 independent derivatives is glucocorticoid-dependent. Cardiovasc Pathol 2019; 46:107194. [PMID: 31982687 DOI: 10.1016/j.carpath.2019.107194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 11/12/2019] [Accepted: 12/10/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Celecoxib, a selective cyclooxygenase-2 inhibitor, was recently associated with increased incidence of aortic stenosis and found to produce a valvular calcification risk in vitro. Several cyclooxygenase-2 independent celecoxib derivatives have been developed and identified as possible therapies for inflammatory diseases due to their cadherin-11 inhibitory functions. Potential cardiovascular toxicities associated with these cyclooxygenase-2 independent celecoxib derivatives have not yet been investigated. Furthermore, the mechanism by which celecoxib produces valvular toxicity is not known. METHODS AND RESULTS Celecoxib treatment produces a 2.8-fold increase in calcification in ex vivo porcine aortic valve leaflets and a more than 2-fold increase in calcification in porcine aortic valve interstitial cells cultured in osteogenic media. Its cyclooxygenase-2 independent derivative, 2,5-dimethylcelecoxib, produces a similar 2.5-fold increase in calcification in ex vivo leaflets and a 13-fold increase in porcine aortic valve interstitial cells cultured in osteogenic media. We elucidate that this offtarget effect depends on the presence of either of the two media components: dexamethasone, a synthetic glucocorticoid used for osteogenic induction, or cortisol, a natural glucocorticoid present at basal levels in the fetal bovine serum. In the absence of glucocorticoids, these inhibitors effectively reduce calcification. By adding glucocorticoids or hydrocortisone to a serum substitute lacking endogenous glucocorticoids, we show that dimethylcelecoxib conditionally induces a 3.5-fold increase in aortic valve calcification and osteogenic expression. Treatment with the Mitogen-activated protein kinase kinase inhibitor, U0126, rescues the offtarget effect, suggesting that celecoxib and dimethylcelecoxib conditionally augment Mitogen-activated protein kinase kinase/extracellular-signal-regulated kinase activity in the presence of glucocorticoids. CONCLUSION Here we identify glucocorticoids as a possible source of the increased valvular calcification risk associated with celecoxib and its cyclooxygenase-2 independent derivatives. In the absence of glucocorticoids, these inhibitors effectively reduce calcification. Furthermore, the offtarget effects are not due to the drug's intrinsic properties as dual cyclooxygenase-2 and cadherin-11 inhibitors. These findings inform future design and development of celecoxib derivatives for potential clinical therapy.
Collapse
|
250
|
Lin Z, Li Z, Li EN, Li X, Del Duke CJ, Shen H, Hao T, O'Donnell B, Bunnell BA, Goodman SB, Alexander PG, Tuan RS, Lin H. Osteochondral Tissue Chip Derived From iPSCs: Modeling OA Pathologies and Testing Drugs. Front Bioeng Biotechnol 2019; 7:411. [PMID: 31921815 PMCID: PMC6930794 DOI: 10.3389/fbioe.2019.00411] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/27/2019] [Indexed: 01/17/2023] Open
Abstract
Osteoarthritis (OA) is a chronic disease mainly characterized by degenerative changes in cartilage, but other joint elements such as bone are also affected. To date, there are no disease-modifying OA drugs (DMOADs), owing in part to a deficiency of current models in simulating OA pathologies and etiologies in humans. In this study, we aimed to develop microphysiological osteochondral (OC) tissue chips derived from human induced pluripotent stem cells (iPSCs) to model the pathologies of OA. We first induced iPSCs into mesenchymal progenitor cells (iMPCs) and optimized the chondro- and osteo-inductive conditions for iMPCs. Then iMPCs were encapsulated into photocrosslinked gelatin scaffolds and cultured within a dual-flow bioreactor, in which the top stream was chondrogenic medium and the bottom stream was osteogenic medium. After 28 days of differentiation, OC tissue chips were successfully generated and phenotypes were confirmed by real time RT-PCR and histology. To create an OA model, interleukin-1β (IL-1β) was used to challenge the cartilage component for 7 days. While under control conditions, the bone tissue promoted chondrogenesis and suppressed chondrocyte terminal differentiation of the overlying chondral tissue. Under conditions modeling OA, the bone tissue accelerated the degradation of chondral tissue which is likely via the production of catabolic and inflammatory cytokines. These findings suggest active functional crosstalk between the bone and cartilage tissue components in the OC tissue chip under both normal and pathologic conditions. Finally, a selective COX-2 inhibitor commonly prescribed drug for OA, Celecoxib, was shown to downregulate the expression of catabolic and proinflammatory cytokines in the OA model, demonstrating the utility of the OC tissue chip model for drug screening. In summary, the iPSC-derived OC tissue chip developed in this study represents a high-throughput platform applicable for modeling OA and for the screening and testing of candidate DMOADs.
Collapse
Affiliation(s)
- Zixuan Lin
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhong Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Eileen N. Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
| | - Xinyu Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
| | - Colin J. Del Duke
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - He Shen
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Tingjun Hao
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
| | - Benjamen O'Donnell
- Department of Pharmacology, Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA, United States
| | - Bruce A. Bunnell
- Department of Pharmacology, Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA, United States
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery and Bioengineering, Stanford University, Stanford, CA, United States
| | - Peter G. Alexander
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Rocky S. Tuan
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Hang Lin
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|