1
|
Naseer N, Mustafa MM, Latief N, Fazal N, Tariq M, Afreen A, Yaqub F, Riazuddin S. Sarcococca saligna fabricated gold nanoparticles alleviated in vitro oxidative stress and inflammation in human adipose-derived stem cells. J Biomed Mater Res B Appl Biomater 2023; 111:2032-2043. [PMID: 37560935 DOI: 10.1002/jbm.b.35303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 06/05/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023]
Abstract
Oxidative stress is a destructive phenomenon that affects various cell structures including membranes, proteins, lipoproteins, lipids, and DNA. Oxidative stress and inflammation owing to lifestyle changes may lead to serious diseases such as Cancers, Gout, and Arthritis etc. These disorders can be prevented using different therapeutic strategies including nanomedicine. Biosynthesized gold nanoparticles (GNPs) because of their anti-inflammatory and antioxidant bioactivities can be key player in reversal of these ailments. This study was carried out to evaluate the anti-inflammatory and antioxidant potential of bio fabricated GNPs with Sarcococca saligna (S. saligna) extract on injured human adipose-derived Mesenchymal stem cells (hADMSCs). GNPs were characterized by ultraviolet-visible (UV-Vis) spectroscopy, Scanning Electron Microscopy (SEM), x-ray diffraction (XRD), Fourier transform infrared spectroscopy (FTIR), and energy dispersive x-ray (EDS). Phytochemical screening of biosynthesized GNPs exhibited a significant release of polyphenols, that is, total phenolic content (TPC) and total flavonoid content (TFC). GNPs priming amended the in vitro injury caused by Monosodium Iodoacetate (MIA) as exhibited by improved cell viability, wound closure response and superoxide dismutase activity (SOD). The anti-inflammatory conduct assessed through NF-κB pathway and other associated inflammatory markers reported down-regulation of TNF-α (0.644 ± 0.045), IL-1β (0.694 ± 0.147) and IL-6 (0.622 ± 0.112), apoptosis causing genes like Caspase-3 (0.734 ± 0.13) and BAX (0.830 ± 0.12), NF-κB pathway, p65 (0.672 ± 0.084) and p105 (0.539 ± 0.083) associated genes. High SOD activity (95 ± 5.25%) revealed by treated hADMSCs with GNPs also supported the antioxidant role of GNPs in vitro model. This study concludes that S. saligna bio fabricated GNPs priming may improve the therapeutic potential of hADMSCs against chronic inflammatory problems by regulating NF-κB pathway.
Collapse
Affiliation(s)
- Nadia Naseer
- Center of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Munam Mustafa
- Center of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Noreen Latief
- Center of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Numan Fazal
- Center of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Tariq
- Department of Biotechnology, Mirpur University of Science and Technology (MUST), Mirpur, Pakistan
| | - Afshan Afreen
- Department of Biotechnology, Mirpur University of Science and Technology (MUST), Mirpur, Pakistan
| | - Faiza Yaqub
- Center of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Sheikh Riazuddin
- Jinnah Burn and Reconstructive Surgery Center, Allama Iqbal Medical College (AIMC), Lahore, Punjab, Pakistan
| |
Collapse
|
2
|
Le Q, Madhu V, Hart JM, Farber CR, Zunder ER, Dighe AS, Cui Q. Current evidence on potential of adipose derived stem cells to enhance bone regeneration and future projection. World J Stem Cells 2021; 13:1248-1277. [PMID: 34630861 PMCID: PMC8474721 DOI: 10.4252/wjsc.v13.i9.1248] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/22/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Injuries to the postnatal skeleton are naturally repaired through successive steps involving specific cell types in a process collectively termed “bone regeneration”. Although complex, bone regeneration occurs through a series of well-orchestrated stages wherein endogenous bone stem cells play a central role. In most situations, bone regeneration is successful; however, there are instances when it fails and creates non-healing injuries or fracture nonunion requiring surgical or therapeutic interventions. Transplantation of adult or mesenchymal stem cells (MSCs) defined by the International Society for Cell and Gene Therapy (ISCT) as CD105+CD90+CD73+CD45-CD34-CD14orCD11b-CD79αorCD19-HLA-DR- is being investigated as an attractive therapy for bone regeneration throughout the world. MSCs isolated from adipose tissue, adipose-derived stem cells (ADSCs), are gaining increasing attention since this is the most abundant source of adult stem cells and the isolation process for ADSCs is straightforward. Currently, there is not a single Food and Drug Administration (FDA) approved ADSCs product for bone regeneration. Although the safety of ADSCs is established from their usage in numerous clinical trials, the bone-forming potential of ADSCs and MSCs, in general, is highly controversial. Growing evidence suggests that the ISCT defined phenotype may not represent bona fide osteoprogenitors. Transplantation of both ADSCs and the CD105- sub-population of ADSCs has been reported to induce bone regeneration. Most notably, cells expressing other markers such as CD146, AlphaV, CD200, PDPN, CD164, CXCR4, and PDGFRα have been shown to represent osteogenic sub-population within ADSCs. Amongst other strategies to improve the bone-forming ability of ADSCs, modulation of VEGF, TGF-β1 and BMP signaling pathways of ADSCs has shown promising results. The U.S. FDA reveals that 73% of Investigational New Drug applications for stem cell-based products rely on CD105 expression as the “positive” marker for adult stem cells. A concerted effort involving the scientific community, clinicians, industries, and regulatory bodies to redefine ADSCs using powerful selection markers and strategies to modulate signaling pathways of ADSCs will speed up the therapeutic use of ADSCs for bone regeneration.
Collapse
Affiliation(s)
- Quang Le
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| | - Vedavathi Madhu
- Orthopaedic Surgery Research, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Joseph M Hart
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| | - Charles R Farber
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, United States
- Departments of Public Health Sciences and Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, United States
| | - Eli R Zunder
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, United States
| | - Abhijit S Dighe
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| | - Quanjun Cui
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, United States
| |
Collapse
|
3
|
Efficacy of chitinase-3-like protein 1 as an in vivo bone formation predictable marker of maxillary/mandibular bone marrow stromal cells. Regen Ther 2021; 18:38-50. [PMID: 33869686 PMCID: PMC8027134 DOI: 10.1016/j.reth.2021.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
Introduction Maxillary/mandibular bone marrow stromal cells (MBMSCs) are a useful cell source for bone regeneration in the oral and maxillofacial region. To further ensure the clinical application of MBMSCs in bone regenerative therapy, it is important to determine the bone formation capacity of MBMSCs before transplantation. The aim of this study is to identify the molecular marker that determines the in vivo bone formation capacity of MBMSCs. Methods The cell growth, cell surface antigens, in vitro and in vivo bone formation capacity of MBMSCs were examined. The amount of chitinase-3-like protein 1 (CHI3L1) secreted into the conditioned medium was quantified. The effects of CHI3L1 on the cell growth and osteogenic differentiation potential of MBMSCs and on the cell growth and migration of vascular endothelial cells and fibroblasts were examined. Results The cell growth, and in vitro and in vivo bone formation capacity of the cells treated with different conditions were observed. MBMSCs that secreted a large amount of CHI3L1 into the conditioned medium tended to have low in vivo bone formation capacity, whereas MBMSCs that secreted a small amount of CHI3L1 had greater in vivo bone formation capacity. CHI3L1 promoted the migration of vascular endothelial cells, and the cell growth and migration of fibroblasts. Conclusion Our study indicates that the in vitro osteogenic differentiation capacity of MBMSCs and the in vivo bone formation capacities of MBMSCs were not necessarily correlated. The transplantation of high CHI3L1 secretory MBMSCs may suppress bone formation by inducing fibrosis at the site. These results suggest that the CHI3L1 secretion levels from MBMSCs may be used as a predictable marker of bone formation capacity in vivo. In vitro and in vivo bone formation capacities of MBMSCs were not correlated. MBMSCs with high CHI3L1 secretion tended to have low in vivo bone formation. MBMSCs with low CHI3L1 secretion tended to have high in vivo bone formation. CHI3L1 can be in vivo bone formation capacity predictable marker of MBMSCs.
Collapse
Key Words
- ALP, Alkaline phosphatase
- BMSC, bone marrow-derived stem cell
- Bone formation capacity
- CHI3L1, chitinase-3-like protein 1
- Chitinase-3-like protein 1
- FBS, fetal bovine serum
- HUVEC, human umbilical vein endothelial cells
- Jaw bone marrow stromal cells
- MBMSC, maxillary/mandibular bone marrow stromal cells
- MSCs, mesenchymal stem cells
- Migration
- NHDF, normal human dermal fibroblasts
- α-MEM, alpha modified Eagle's minimum essential medium
- β-TCP, beta-tricalcium phosphate
Collapse
|
4
|
Sun Q, Nakata H, Yamamoto M, Kasugai S, Kuroda S. Comparison of gingiva-derived and bone marrow mesenchymal stem cells for osteogenesis. J Cell Mol Med 2019; 23:7592-7601. [PMID: 31657140 PMCID: PMC6815943 DOI: 10.1111/jcmm.14632] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 07/15/2019] [Accepted: 08/06/2019] [Indexed: 01/01/2023] Open
Abstract
Presently, bone marrow is considered as a prime source of mesenchymal stem cells; however, there are some drawbacks and limitations. Compared with other mesenchymal stem cell (MSC) sources, gingiva-derived mesenchymal stem cells (GMSCs) are abundant and easy to obtain through minimally invasive cell isolation techniques. In this study, MSCs derived from gingiva and bone marrow were isolated and cultured from mice. GMSCs were characterized by osteogenic, adipogenic and chondrogenic differentiation, and flow cytometry. Compared with bone marrow MSCs (BMSCs), the proliferation capacity was judged by CCK-8 proliferation assay. Osteogenic differentiation was assessed by ALP staining, ALP assay and Alizarin red staining. RT-qPCR was performed for ALP, OCN, OSX and Runx2. The results indicated that GMSCs showed higher proliferative capacity than BMSCs. GMSCs turned more positive for ALP and formed a more number of mineralized nodules than BMSCs after osteogenic induction. RT-qPCR revealed that the expression of ALP, OCN, OSX and Runx2 was significantly increased in the GMSCs compared with that in BMSCs. Moreover, it was found that the number of CD90-positive cells in GMSCs elevated more than that of BMSCs during osteogenic induction. Taking these results together, it was indicated that GMSCs might be a promising source in the future bone tissue engineering.
Collapse
Affiliation(s)
- Quan Sun
- Department of Oral Implantology and Regenerative Dental Medicine, Division of Oral Health Sciences, Medical and Dental Sciences Track, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Hidemi Nakata
- Department of Oral Implantology and Regenerative Dental Medicine, Division of Oral Health Sciences, Medical and Dental Sciences Track, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Maiko Yamamoto
- Department of Oral Implantology and Regenerative Dental Medicine, Division of Oral Health Sciences, Medical and Dental Sciences Track, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Shohei Kasugai
- Department of Oral Implantology and Regenerative Dental Medicine, Division of Oral Health Sciences, Medical and Dental Sciences Track, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Shinji Kuroda
- Department of Oral Implantology and Regenerative Dental Medicine, Division of Oral Health Sciences, Medical and Dental Sciences Track, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| |
Collapse
|
5
|
Born S, Dörfel MJ, Hartjen P, Haschemi Yekani SA, Luecke J, Meutsch JK, Westphal JK, Birkelbach M, Köhnke R, Smeets R, Krueger M. A short-term plastic adherence incubation of the stromal vascular fraction leads to a predictable GMP-compliant cell-product. ACTA ACUST UNITED AC 2019; 9:161-172. [PMID: 31508331 PMCID: PMC6726751 DOI: 10.15171/bi.2019.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/14/2022]
Abstract
![]()
Introduction:Mesenchymal stromal/stem cells (MSCs) derived from fat tissue are an encouraging tool for regenerative medicine. They share properties similar to the bone marrow-derived MSCs, but the amount of MSCs per gram of fat tissue is 500x higher. The fat tissue can easily be digested by collagenase, releasing a heterogeneous cell fraction called stromal vascular fraction (SVF) which contains a variable amount of stromal/stem cells. In Europe, cell products like the SVF derived from fat tissue are considered advanced therapy medicinal product (ATMPs). As a consequence, the manufacturing process has to be approved via GMP-compliant process validation. The problem of the process validation for SVF is the heterogeneity of this fraction.
Methods: Here, we modified existing purification strategies by adding an additional plastic adherence incubation of maximal 20 hours after SVF isolation. The resulting cell fraction was characterized and compared to SVF as well as cultivated adipose-derived stromal/stem cells (ASCs) with respect to viability and cell yield, the expression of surface markers, differentiation potential and cytokine expression.
Results: Short-term incubation significantly reduced the heterogeneity of the resulting cell fraction compared to SVF. The cells were able to differentiate into adipocytes, chondrocytes, and osteoblasts. More importantly, they expressed trophic proteins which have been previously associated with the beneficial effects of MSCs. Furthermore, GMP compliance of the production process described herein was acknowledged by the national regulatory agencies (DE_BB_01_GMP_2017_1018).
Conclusion: Addition of a short purification-step after the SVF isolation is a cheap and fast strategy to isolate a homogeneous uncultivated GMP-compliant cell fraction of ASCs.
Collapse
Affiliation(s)
| | | | - Philip Hartjen
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | - Moritz Birkelbach
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Robert Köhnke
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany.,Division, Regenerative Orofacial Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
6
|
Saalbach A, Anderegg U. Thy‐1: more than a marker for mesenchymal stromal cells. FASEB J 2019; 33:6689-6696. [DOI: 10.1096/fj.201802224r] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Anja Saalbach
- Department of Dermatology, Venerology, and AllergologyFaculty of MedicineLeipzig UniversityLeipzigGermany
| | - Ulf Anderegg
- Department of Dermatology, Venerology, and AllergologyFaculty of MedicineLeipzig UniversityLeipzigGermany
| |
Collapse
|
7
|
Uniaxial Cyclic Tensile Stretching at 8% Strain Exclusively Promotes Tenogenic Differentiation of Human Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells Int 2019; 2019:9723025. [PMID: 30918524 PMCID: PMC6409073 DOI: 10.1155/2019/9723025] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/13/2018] [Accepted: 11/08/2018] [Indexed: 12/18/2022] Open
Abstract
The present study was conducted to establish the amount of mechanical strain (uniaxial cyclic stretching) required to provide optimal tenogenic differentiation expression in human mesenchymal stromal cells (hMSCs) in vitro, in view of its potential application for tendon maintenance and regeneration. Methods. In the present study, hMSCs were subjected to 1 Hz uniaxial cyclic stretching for 6, 24, 48, and 72 hours; and were compared to unstretched cells. Changes in cell morphology were observed under light and atomic force microscopy. The tenogenic, osteogenic, adipogenic, and chondrogenic differentiation potential of hMSCs were evaluated using biochemical assays, extracellular matrix expressions, and selected mesenchyme gene expression markers; and were compared to primary tenocytes. Results. Cells subjected to loading displayed cytoskeletal coarsening, longer actin stress fiber, and higher cell stiffness as early as 6 hours. At 8% and 12% strains, an increase in collagen I, collagen III, fibronectin, and N-cadherin production was observed. Tenogenic gene expressions were highly expressed (p < 0.05) at 8% (highest) and 12%, both comparable to tenocytes. In contrast, the osteoblastic, chondrogenic, and adipogenic marker genes appeared to be downregulated. Conclusion. Our study suggests that mechanical loading at 8% strain and 1 Hz provides exclusive tenogenic differentiation; and produced comparable protein and gene expression to primary tenocytes.
Collapse
|
8
|
Li R, Lin QX, Liang XZ, Liu GB, Tang H, Wang Y, Lu SB, Peng J. Stem cell therapy for treating osteonecrosis of the femoral head: From clinical applications to related basic research. Stem Cell Res Ther 2018; 9:291. [PMID: 30359305 PMCID: PMC6202807 DOI: 10.1186/s13287-018-1018-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a refractory disease that is associated with collapse of the femoral head, with a risk of hip arthroplasty in younger populations. Thus, there has been an increased focus on early interventions for ONFH that aim to preserve the native articulation. Stem cell therapy is a promising treatment, and an increasing number of recent studies have focused on this topic. Many clinical studies have reported positive outcomes of stem cell therapy for the treatment of ONFH. To improve the therapeutic effects of this approach, many related basic research studies have also been performed. However, some issues must be further explored, such as the appropriate patient selection procedure, the optimal stem cell selection protocol, the ideal injection number, and the safety of stem cell therapy. The purpose of this review is to summarize the available clinical studies and basic research related to stem cell therapy for ONFH.
Collapse
Affiliation(s)
- Rui Li
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Qiu-Xia Lin
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Xue-Zhen Liang
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355 Shandong China
| | - Guang-Bo Liu
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - He Tang
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Yu Wang
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Shi-Bi Lu
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| | - Jiang Peng
- Institute of Orthopedics, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, 100853 China
| |
Collapse
|
9
|
Olimpio RMC, de Oliveira M, De Sibio MT, Moretto FCF, Deprá IC, Mathias LS, Gonçalves BM, Rodrigues BM, Tilli HP, Coscrato VE, Costa SMB, Mazeto GMFS, Fernandes CJC, Zambuzzi WF, Saraiva PP, Maria DA, Nogueira CR. Cell viability assessed in a reproducible model of human osteoblasts derived from human adipose-derived stem cells. PLoS One 2018; 13:e0194847. [PMID: 29641603 PMCID: PMC5895002 DOI: 10.1371/journal.pone.0194847] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/09/2018] [Indexed: 01/04/2023] Open
Abstract
Human adipose tissue-derived stem cells (hASCs) have been subjected to extensive investigation because of their self-renewal properties and potential to restore damaged tissues. In the literature, there are several protocols for differentiating hASCs into osteoblasts, but there is no report on the control of cell viability during this process. In this study, we used osteoblasts derived from hASCs of patients undergoing abdominoplasty. The cells were observed at the beginning and end of bone matrix formation, and the expression of proteins involved in this process, including alkaline phosphatase and osteocalcin, was assessed. RANKL, Osterix, Runx2, Collagen3A1, Osteopontin and BSP expression levels were analyzed using real-time PCR, in addition to a quantitative assessment of protein levels of the markers CD45, CD105, STRO-1, and Nanog, using immunofluorescence. Rhodamine (Rho123), cytochrome-c, caspase-3, P-27, cyclin D1, and autophagy cell markers were analyzed by flow cytometry to demonstrate potential cellular activity and the absence of apoptotic and tumor cell processes before and after cell differentiation. The formation of bone matrix, along with calcium nodules, was observed after 16 days of osteoinduction. The gene expression levels of RANKL, Osterix, Runx2, Collagen3A1, Osteopontin, BSP and alkaline phosphatase activity were also elevated after 16 days of osteoinduction, whereas the level of osteocalcin was higher after 21 days of osteoinduction. Our data also showed that the cells had a high mitochondrial membrane potential and a low expression of apoptotic and tumor markers, both before and after differentiation. Cells were viable after the different phases of differentiation. This proposed methodology, using markers to evaluate cell viability, is therefore successful in assessing different phases of stem cell isolation and differentiation.
Collapse
Affiliation(s)
- Regiane M. C. Olimpio
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
- * E-mail:
| | - Miriane de Oliveira
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Maria T. De Sibio
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Fernanda C. F. Moretto
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Igor C. Deprá
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Lucas S. Mathias
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Bianca M. Gonçalves
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Bruna M. Rodrigues
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Helena P. Tilli
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Virgínia E. Coscrato
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Sarah M. B. Costa
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Gláucia M. F. S. Mazeto
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Célio J. C. Fernandes
- Institute of Biosciences, Department of Chemistry and Biochemistry, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Willian F. Zambuzzi
- Institute of Biosciences, Department of Chemistry and Biochemistry, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Patrícia P. Saraiva
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Durvanei A. Maria
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, São Paulo, Brazil
| | - Célia R. Nogueira
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| |
Collapse
|
10
|
Itoh H, Nishikawa S, Haraguchi T, Arikawa Y, Eto S, Hiyama M, Iseri T, Itoh Y, Nakaichi M, Sakai Y, Tani K, Taura Y, Itamoto K. Aldehyde dehydrogenase activity helps identify a subpopulation of murine adipose-derived stem cells with enhanced adipogenic and osteogenic differentiation potential. World J Stem Cells 2017; 9:179-186. [PMID: 29104736 PMCID: PMC5661130 DOI: 10.4252/wjsc.v9.i10.179] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/11/2017] [Accepted: 09/13/2017] [Indexed: 02/07/2023] Open
Abstract
AIM To identify and characterize functionally distinct subpopulation of adipose-derived stem cells (ADSCs).
METHODS ADSCs cultured from mouse subcutaneous adipose tissue were sorted fluorescence-activated cell sorter based on aldehyde dehydrogenase (ALDH) activity, a widely used stem cell marker. Differentiation potentials were analyzed by utilizing immunocytofluorescece and its quantitative analysis.
RESULTS Approximately 15% of bulk ADSCs showed high ALDH activity in flow cytometric analysis. Although significant difference was not seen in proliferation capacity, the adipogenic and osteogenic differentiation capacity was higher in ALDHHi subpopulations than in ALDHLo. Gene set enrichment analysis revealed that ribosome-related gene sets were enriched in the ALDHHi subpopulation.
CONCLUSION High ALDH activity is a useful marker for identifying functionally different subpopulations in murine ADSCs. Additionally, we suggested the importance of ribosome for differentiation of ADSCs by gene set enrichment analysis.
Collapse
Affiliation(s)
- Harumichi Itoh
- Department of Small Animal Clinical Science, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Shimpei Nishikawa
- Department of Small Animal Clinical Science, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Tomoya Haraguchi
- Department of Small Animal Clinical Science, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Yu Arikawa
- Department of Small Animal Clinical Science, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Shotaro Eto
- Department of Veterinary Surgery, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Masato Hiyama
- Department of Veterinary Surgery, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Toshie Iseri
- Department of Veterinary Radiology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Yoshiki Itoh
- Department of Veterinary Radiology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Munekazu Nakaichi
- Department of Veterinary Radiology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Yusuke Sakai
- Department of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Kenji Tani
- Department of Veterinary Surgery, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Yasuho Taura
- Department of Veterinary Surgery, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| | - Kazuhito Itamoto
- Department of Small Animal Clinical Science, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-8511, Japan
| |
Collapse
|
11
|
Bearden RN, Huggins SS, Cummings KJ, Smith R, Gregory CA, Saunders WB. In-vitro characterization of canine multipotent stromal cells isolated from synovium, bone marrow, and adipose tissue: a donor-matched comparative study. Stem Cell Res Ther 2017; 8:218. [PMID: 28974260 PMCID: PMC5627404 DOI: 10.1186/s13287-017-0639-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 07/06/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022] Open
Abstract
Background The dog represents an excellent large animal model for translational cell-based studies. Importantly, the properties of canine multipotent stromal cells (cMSCs) and the ideal tissue source for specific translational studies have yet to be established. The aim of this study was to characterize cMSCs derived from synovium, bone marrow, and adipose tissue using a donor-matched study design and a comprehensive series of in-vitro characterization, differentiation, and immunomodulation assays. Methods Canine MSCs were isolated from five dogs with cranial cruciate ligament rupture. All 15 cMSC preparations were evaluated using colony forming unit (CFU) assays, flow cytometry analysis, RT-PCR for pluripotency-associated genes, proliferation assays, trilineage differentiation assays, and immunomodulation assays. Data were reported as mean ± standard deviation and compared using repeated-measures analysis of variance and Tukey post-hoc test. Significance was established at p < 0.05. Results All tissue samples produced plastic adherent, spindle-shaped preparations of cMSCs. Cells were negative for CD34, CD45, and STRO-1 and positive for CD9, CD44, and CD90, whereas the degree to which cells were positive for CD105 was variable depending on tissue of origin. Cells were positive for the pluripotency-associated genes NANOG, OCT4, and SOX2. Accounting for donor and tissue sources, there were significant differences in CFU potential, rate of proliferation, trilineage differentiation, and immunomodulatory response. Synovium and marrow cMSCs exhibited superior early osteogenic activity, but when assessing late-stage osteogenesis no significant differences were detected. Interestingly, bone morphogenic protein-2 (BMP-2) supplementation was necessary for early-stage and late-stage osteogenic differentiation, a finding consistent with other canine studies. Additionally, synovium and adipose cMSCs proliferated more rapidly, displayed higher CFU potential, and formed larger aggregates in chondrogenic assays, although proteoglycan and collagen type II staining were subjectively decreased in adipose pellets as compared to synovial and marrow pellets. Lastly, cMSCs derived from all three tissue sources modulated murine macrophage TNF-α and IL-6 levels in a lipopolysaccharide-stimulated coculture assay. Conclusions While cMSCs from synovium, marrow, and adipose tissue share a number of similarities, important differences in proliferation and trilineage differentiation exist and should be considered when selecting cMSCs for translational studies. These results and associated methods will prove useful for future translational studies involving the canine model. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0639-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robert N Bearden
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Shannon S Huggins
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Kevin J Cummings
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Roger Smith
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Carl A Gregory
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, College of Medicine, Texas A&M University, College Station, TX, USA
| | - William B Saunders
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
12
|
Techatharatip O, Nowwarote N, Taebunpakul S, Pavasant P. Biphasic Effect of ATP on In Vitro Mineralization of Dental Pulp Cells. J Cell Biochem 2017; 119:488-498. [PMID: 28598038 DOI: 10.1002/jcb.26206] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/08/2017] [Indexed: 01/01/2023]
Abstract
Dental pulp cells release adenosine triphosphate (ATP) in response to intrapulpal pressure and the amount released depends on the magnitude of the pressure. ATP regulates the differentiation of stem cells into adipocytes and osteoblasts. However, it is unknown whether extracellular ATP influences the stemness and osteogenic differentiation of stem cells from human exfoliated deciduous teeth (SHEDs). Therefore, this study investigated the effects of extracellular ATP at a low (0.1 μM) and high (10 μM) concentration on the stemness and osteogenic differentiation of SHEDs. Cells were cultured in either growth medium or osteogenic medium with or without 0.1-10 μM ATP. In growth medium, both concentrations of ATP increased the mRNA expression of pluripotent and osteogenic markers. In contrast, in osteogenic medium, 0.1 μM ATP enhanced in vitro mineralization, whereas 10 μM ATP inhibited this process. In addition, 10 μM ATP stimulated the mRNA expression and activity of ectonucleotide pyrophosphatase/phosphodiesterase (ENPP), an enzyme that regulates the phosphate/pyrophosphate ratio. Thus, depending on the growth condition and its concentration, ATP stimulated stemness and in vitro mineralization or inhibited mineralization. In growth medium, both ATP concentrations stimulated pluripotent and osteogenic marker gene expression. However, in osteogenic medium, a biphasic effect was found on in vitro mineralization; the low concentration stimulated, whereas the high concentration inhibited, mineralization. We propose that ATP released due to mechanical stress modulates the stemness and differentiation of SHEDs. J. Cell. Biochem. 119: 488-498, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Oranuch Techatharatip
- Graduate Program in Oral Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Nunthawan Nowwarote
- Graduate Program in Oral Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | | | - Prasit Pavasant
- Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Faculty of Dentistry, Department of Anatomy, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
13
|
Brett E, Flacco J, Blackshear C, Longaker MT, Wan DC. Biomimetics of Bone Implants: The Regenerative Road. Biores Open Access 2017; 6:1-6. [PMID: 28163982 PMCID: PMC5248549 DOI: 10.1089/biores.2016.0044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The current strategies for healing bone defects are numerous and varied. At the core of each bone healing therapy is a biomimetic mechanism, which works to enhance bone growth. These range from porous scaffolds, bone mineral usage, collagen, and glycosaminoglycan substitutes to transplanted cell populations. Bone defects face a range of difficulty in their healing, given the composite of dense outer compact bone and blood-rich inner trabecular bone. As such, the tissue possesses a number of inherent characteristics, which may be clinically harnessed as promoters of bone healing. These include mechanical characteristics, mineral composition, native collagen content, and cellular fraction of bone. This review charts multiple biomimetic strategies to help heal bony defects in large and small osseous injury sites, with a special focus on cell transplantation.
Collapse
Affiliation(s)
- Elizabeth Brett
- Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - John Flacco
- Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Charles Blackshear
- Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine, Stanford, California.; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
14
|
Evaluation of 2 Purification Methods for Isolation of Human Adipose-Derived Stem Cells Based on Red Blood Cell Lysis With Ammonium Chloride and Hypotonic Sodium Chloride Solution. Ann Plast Surg 2017; 78:83-90. [DOI: 10.1097/sap.0000000000000953] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Gomathysankar S, Halim AS, Yaacob NS, Noor NM, Mohamed M. Compatibility of Porous Chitosan Scaffold with the Attachment and Proliferation of human Adipose-Derived Stem Cells In Vitro. J Stem Cells Regen Med 2016. [PMID: 28096632 PMCID: PMC5227107 DOI: 10.46582/jsrm.1202012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adipose-derived stem cells (ASCs) have potential applications in the repair and regeneration of various tissues and organs. The use of various scaffold materials as an excellent template for mimicking the extracellular matrix to induce the attachment and proliferation of different cell types has always been of interest in the field of tissue engineering because ideal biomaterials are in great demand. Chitosan, a marine polysaccharide, have wide clinical applications and it acts as a promising scaffold for cell migration and proliferation. ASCs, with their multi-differentiation potential, and chitosan, with its great biocompatibility with ASCs, were investigated in the present study. ASCs were isolated and were characterized by two different methods: immunocytochemistry and flow cytometry, using the mesenchymal stem cell markers CD90, CD105, CD73 and CD29. The ASCs were then induced to differentiate into adipogenic, osteogenic and chondrogenic lineages. These ASCs were incorporated into a porous chitosan scaffold (PCS), and their structural morphology was studied using a scanning electron microscope and hematoxylin and eosin staining. The proliferation rate of the ASCs on the PCS was assessed using a PrestoBlue viability assay. The results indicated that the PCS provides an excellent template for the adhesion and proliferation of ASCs. Thus, this study revealed that PCS is a promising biomaterial for inducing the proliferation of ASCs, which could lead to successful tissue reconstruction in the field of tissue engineering.
Collapse
Affiliation(s)
- Sankaralakshmi Gomathysankar
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Ahmad Sukari Halim
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nik Soriani Yaacob
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang kerian, Kelantan, Malaysia
| | - Norhayati Mohd Noor
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Mohaini Mohamed
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
16
|
Gomathysankar S, Halim AS, Yaacob NS, Noor NM, Mohamed M. Compatibility of Porous Chitosan Scaffold with the Attachment and Proliferation of human Adipose-Derived Stem Cells In Vitro. J Stem Cells Regen Med 2016; 12:79-86. [PMID: 28096632 PMCID: PMC5227107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 07/11/2016] [Indexed: 11/13/2023]
Abstract
Adipose-derived stem cells (ASCs) have potential applications in the repair and regeneration of various tissues and organs. The use of various scaffold materials as an excellent template for mimicking the extracellular matrix to induce the attachment and proliferation of different cell types has always been of interest in the field of tissue engineering because ideal biomaterials are in great demand. Chitosan, a marine polysaccharide, have wide clinical applications and it acts as a promising scaffold for cell migration and proliferation. ASCs, with their multi-differentiation potential, and chitosan, with its great biocompatibility with ASCs, were investigated in the present study. ASCs were isolated and were characterized by two different methods: immunocytochemistry and flow cytometry, using the mesenchymal stem cell markers CD90, CD105, CD73 and CD29. The ASCs were then induced to differentiate into adipogenic, osteogenic and chondrogenic lineages. These ASCs were incorporated into a porous chitosan scaffold (PCS), and their structural morphology was studied using a scanning electron microscope and hematoxylin and eosin staining. The proliferation rate of the ASCs on the PCS was assessed using a PrestoBlue viability assay. The results indicated that the PCS provides an excellent template for the adhesion and proliferation of ASCs. Thus, this study revealed that PCS is a promising biomaterial for inducing the proliferation of ASCs, which could lead to successful tissue reconstruction in the field of tissue engineering.
Collapse
Affiliation(s)
- Sankaralakshmi Gomathysankar
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Ahmad Sukari Halim
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nik Soriani Yaacob
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang kerian, Kelantan, Malaysia.
| | - Norhayati Mohd Noor
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Mohaini Mohamed
- Reconstructive Sciences Unit, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
17
|
Understanding the structural features of symptomatic calcific aortic valve stenosis: A broad-spectrum clinico-pathologic study in 236 consecutive surgical cases. Int J Cardiol 2016; 228:364-374. [PMID: 27866029 DOI: 10.1016/j.ijcard.2016.11.180] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 11/06/2016] [Indexed: 11/20/2022]
Abstract
BACKGROUND With age, aortic valve cusps undergo varying degrees of sclerosis which, sometimes, can progress to calcific aortic valve stenosis (AVS). To perform a retrospective clinico-pathologic investigation in patients with calcific AVS. METHODS We characterized and graded the structural remodeling in 236 aortic valves (200 tricuspid and 36 bicuspid) from patients with calcific AVS (148 males; average 72years); possible relationships between general/clinical/echocardiographic characteristics and the histopathologic changes were explored. Twenty autopsy aortic valves served as controls. In 40 cases, we also tested the immunohistochemical expression of metalloproteinases and cytokines, and characterized the inflammatory infiltrate. In 5 cases, we cultured cusp stem cells and explored their potential to differentiate into osteoblasts/adipocytes. RESULTS AVS cusps showed structural remodeling as severe fibrosis (100%), calcific nodules (100%), neoangiogenesis (81%), inflammation (71%), bone metaplasia with or without hematopoiesis (6% and 53%, respectively), adipose metaplasia (16%), and cartilaginous metaplasia (7%). At multivariate analysis, AVS degree and interventricular septum thickness were the only predictors of remodeling (barring inflammation). All the tested metalloproteinases (except MMP-13) and cytokines were expressed in AVS cusps. Inflammation mainly consisted of B and T lymphocytes (CD4+/CD8+ cell ratio 3:1) and plasma cells. AVS changes were mostly different from typical atherosclerosis. Cultured mesenchymal cusp stem cells could differentiate into osteoblasts/adipocytes. CONCLUSIONS Structural remodeling in AVS is peculiar and considerable, and is related to the severity of the disease. However, the different newly formed tissues-where "valvular interstitial cells" play a key role-and their well-known slow turnover suggest a reverse structural remodeling improbable.
Collapse
|
18
|
Mesenchymal Stem Cells Subpopulations: Application for Orthopedic Regenerative Medicine. Stem Cells Int 2016; 2016:3187491. [PMID: 27725838 PMCID: PMC5048051 DOI: 10.1155/2016/3187491] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/10/2016] [Accepted: 08/07/2016] [Indexed: 12/21/2022] Open
Abstract
Research on mesenchymal stem cells (MSCs) continues to progress rapidly. Nevertheless, the field faces several challenges, such as inherent cell heterogeneity and the absence of unique MSCs markers. Due to MSCs' ability to differentiate into multiple tissues, these cells represent a promising tool for new cell-based therapies. However, for tissue engineering applications, it is critical to start with a well-defined cell population. Additionally, evidence that MSCs subpopulations may also feature distinct characteristics and regeneration potential has arisen. In this report, we present an overview of the identification of MSCs based on the expression of several surface markers and their current tissue sources. We review the use of MSCs subpopulations in recent years and the main methodologies that have addressed their isolation, and we emphasize the most-used surface markers for selection, isolation, and characterization. Next, we discuss the osteogenic and chondrogenic differentiation from MSCs subpopulations. We conclude that MSCs subpopulation selection is not a minor concern because each subpopulation has particular potential for promoting the differentiation into osteoblasts and chondrocytes. The accurate selection of the subpopulation advances possibilities suitable for preclinical and clinical studies and determines the safest and most efficacious regeneration process.
Collapse
|
19
|
Semaphorin 3A Shifts Adipose Mesenchymal Stem Cells towards Osteogenic Phenotype and Promotes Bone Regeneration In Vivo. Stem Cells Int 2016; 2016:2545214. [PMID: 27721834 PMCID: PMC5046026 DOI: 10.1155/2016/2545214] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 01/08/2023] Open
Abstract
Adipose mesenchymal stem cells (ASCs) are considered as the promising seed cells for bone regeneration. However, the lower osteogenic differentiation capacity limits its therapeutic efficacy. Identification of the key molecules governing the differences between ASCs and BMSCs would shed light on manipulation of ASCs towards osteogenic phenotype. In this study, we screened semaphorin family members in ASCs and BMSCs and identified Sema3A as an osteogenic semaphorin that was significantly and predominantly expressed in BMSCs. The analyses in vitro showed that the overexpression of Sema3A in ASCs significantly enhanced the expression of bone-related genes and extracellular matrix calcium deposition, while decreasing the expression of adipose-related genes and thus lipid droplet formation, resembling a BMSCs phenotype. Furthermore, Sema3A modified ASCs were then engrafted into poly(lactic-co-glycolic acid) (PLGA) scaffolds to repair the critical-sized calvarial defects in rat model. As expected, Sema3A modified ASCs encapsulation significantly promoted new bone formation with higher bone volume fraction and bone mineral density. Additionally, Sema3A was found to simultaneously increase multiple Wnt related genes and thus activating Wnt pathway. Taken together, our study here identifies Sema3A as a critical gene for osteogenic phenotype and reveals that Sema3A-modified ASCs would serve as a promising candidate for bettering bone defect repair.
Collapse
|
20
|
Song X, Hong C, Zheng Q, Zhao H, Song K, Liu Z, Shen J, Li Y, Wang J, Shen T. Differentiation potential of rabbit CD90-positive cells sorted from adipose-derived stem cells in vitro. In Vitro Cell Dev Biol Anim 2016; 53:77-82. [PMID: 27553254 DOI: 10.1007/s11626-016-0081-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/26/2016] [Indexed: 01/15/2023]
Abstract
To investigate the differentiation potential of purified CD90+ cells sorted from adipose-derived stem cells (ADSCs), CD90+ cells were sorted from rabbit ADSCs using flow cytometry. Then, cell expansion of CD90+ cells and unsorted ADSCs was observed using an inverted microscope. Furthermore, cell surface markers including CD40, CD105, and CD90 on CD90+ cells and unsorted ADSCs were quantified using flow cytometry. Additionally, multi-lineage differentiation ability between CD90+ cells and unsorted ADSCs was compared, and expression of adipocyte-related genes PPAR-r and CEBPA as well as stem cell-related gene SOX2 in CD90+ cells and unsorted ADSCs was determined using real-time quantitative PCR. We found that CD90+ cells had a stronger cell proliferation ability than unsorted ADSCs. CD90+ cells showed a stronger ability of osteoblast and chondrocyte differentiation than unsorted ADSCs and CD90- cells, whereas the adipose differentiation ability of CD90+ cells was similar to that of ADSCs and CD90- cells. CD14, CD105, and CD90 on CD90+ cells were expressed more highly than those on ADSCs. Additionally, the mRNA expression level of SOX2 in CD90+ cells was significantly higher than that in ADSCs, whereas the expression of PPAR-r and CEBPA was markedly lower than that in ADSCs. These results suggested that the purified CD90+ cells sorted from ADSCs exhibit a stronger differentiation potential than the unsorted ADSCs.
Collapse
Affiliation(s)
- Xinghui Song
- Facility for Biochemistry and Molecular medicine Core Facilities, Zhejiang University School of Medicine, 866 Yuhangtang Rd, Hangzhou, Zhejiang, 310058, China
| | - Chaoyang Hong
- Department of ophthalmology, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Qingqing Zheng
- Department of ophthalmology, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Hailan Zhao
- Department of ophthalmology, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Kangping Song
- Stevenson school, 3152 Forest Lake Road, Pebble Beach, CA, 93953, USA
| | - Zhe Liu
- Department of ophthalmology, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Jiang Shen
- Department of ophthalmology, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Yanwei Li
- Facility for Biochemistry and Molecular medicine Core Facilities, Zhejiang University School of Medicine, 866 Yuhangtang Rd, Hangzhou, Zhejiang, 310058, China
| | - Jiajia Wang
- Facility for Biochemistry and Molecular medicine Core Facilities, Zhejiang University School of Medicine, 866 Yuhangtang Rd, Hangzhou, Zhejiang, 310058, China
| | - Ting Shen
- Department of ophthalmology, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, People's Republic of China.
| |
Collapse
|
21
|
Kim YK, Nakata H, Yamamoto M, Miyasaka M, Kasugai S, Kuroda S. Osteogenic Potential of Mouse Periosteum-Derived Cells Sorted for CD90 In Vitro and In Vivo. Stem Cells Transl Med 2015; 5:227-34. [PMID: 26718647 DOI: 10.5966/sctm.2015-0013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 08/13/2015] [Indexed: 12/19/2022] Open
Abstract
The treatment of bone defects still presents complex problems, although various techniques have been developed. The periosteum is considered a good source of osteogenic precursor cells for new bone formation. It can be collected easily in the clinical setting and is less invasive to the donor site. However, the murine skull periosteum has a poor cellular component, and growth is very slow, making it important to identify a culture method for efficient growth. In the present study, we used three-dimensional cell migration with atelocollagen and gelatin media and found that both were effective for promoting the proliferation of periosteum-derived cells. Moreover, atelocollagen medium is expected to provide an added benefit as a scaffold structure in the ambient temperature of the human body. The selection of a proper surface marker for osteogenesis is imperative for bone regeneration. CD90 is a mesenchymal stem cell marker. Periosteum-derived cells sorted with CD90 showed higher proliferative capacity and osteogenic potential than that of unsorted periosteum-derived cells in vivo and in vitro. Thus, periosteum-derived cells sorted with CD90 are expected to be a good source for bone regeneration. Significance: Periosteum-derived cells showed higher proliferative capacity and osteogenic potential. Periosteum can be collected easily in the clinical setting and is less invasive to the donor site. Thus, periosteum-derived cells can be expected to be a good source for bone regeneration.
Collapse
Affiliation(s)
- You-Kyoung Kim
- Department of Oral Implantology and Regenerative Dental Medicine, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hidemi Nakata
- Department of Oral Implantology and Regenerative Dental Medicine, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Maiko Yamamoto
- Department of Oral Implantology and Regenerative Dental Medicine, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Munemitsu Miyasaka
- Department of Oral Implantology and Regenerative Dental Medicine, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shohei Kasugai
- Department of Oral Implantology and Regenerative Dental Medicine, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Kuroda
- Department of Oral Implantology and Regenerative Dental Medicine, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
22
|
Mohammadpour H, Pourfathollah AA, Nikougoftar Zarif M, Hashemi SM. Increasing proliferation of murine adipose tissue-derived mesenchymal stem cells by TNF-α plus IFN-γ. Immunopharmacol Immunotoxicol 2015; 38:68-76. [DOI: 10.3109/08923973.2015.1115519] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|