1
|
Tang Y, Hu Y, Ding X, Luo D, Li C, Daraqel B, Zheng L. Enriched H3K27Me3 on BMP4 suppresses the osteoblastic differentiation potential of BMSCs in diabetes mellitus. Biochem Biophys Res Commun 2024; 735:150741. [PMID: 39401480 DOI: 10.1016/j.bbrc.2024.150741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 11/05/2024]
Abstract
Diabetes mellitus has been widely acknowledged to have a negative effect on the osteoblastic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). However, the underlying epigenetic mechanisms associated with this process remain to be elucidated. The goal of the present study was to investigate the effect of diabetes mellitus on the osteoblastic differentiation of BMSCs and assess the role of histone methylation in the observed phenomena. The osteoblastic differentiation ability of BMSCs was shown to be decreased in diabetes mellitus, as indicated by alkaline phosphatase activity and the mRNA levels of osteoblast-related genes. Furthermore, diabetes mellitus caused an increased expression of the histone methylase EZH2 and the levels of H3K27Me3 and decreased the expression of the histone demethylase KDM6B, as demonstrated by qRT-PCR and western blotting. Furthermore, immunofluorescence staining suggested that both EZH2 and H3K27Me3 were primarily localized in the nucleus. In addition, chromatin immunoprecipitation assays indicated an increased presence of H3K27Me3 on the promoter region of the BMP4 gene. In summary, in the present study, we demonstrated that the osteoblastic differentiation of BMSCs is dramatically reduced in diabetes mellitus. In addition, upregulation of EZH2 expression and downregulation of KDM6B expression may not be enough to eliminate transcriptional repression mediated by H3K27Me3 on the promoter region of the BMP4 gene during the osteoblastic differentiation of BMSCs in diabetes mellitus.
Collapse
Affiliation(s)
- Yu Tang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China; Chongqing the Seventh People's Hospital, Chongqing, 400054, China
| | - Yun Hu
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Xiaoqian Ding
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Dan Luo
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Caiyu Li
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Baraa Daraqel
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Leilei Zheng
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China; Laboratory of Medical Biochemistry, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China.
| |
Collapse
|
2
|
Song M, Deng M, Peng Z, Dai F, Wang Y, Shu W, Zhou X, Zhang J, Hou Y, Yu B. Granulocyte colony-stimulating factor mediates bone loss via the activation of IL-1β/JNK signaling pathway in murine Staphylococcus aureus-induced osteomyelitis. Int Immunopharmacol 2024; 141:112959. [PMID: 39163688 DOI: 10.1016/j.intimp.2024.112959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024]
Abstract
Staphylococcus aureus (S. aureus)-induced bone loss is a significant challenge in the treatment of osteomyelitis. Our previous study was the first to confirm that granulocyte colony-stimulating factor (G-CSF) mediates S. aureus-induced bone loss. However, the underlying mechanism remains unknown. The objective of this study was to elucidate this. We found G-CSF mediated BMSC senescence and increased IL-1β concentration of serum and bone marrow in mice after S. aureus infection. Furthermore, we demonstrated that G-CSF promoted the expression of IL1b in murine bone marrow-derived neutrophils. Notably, we identified that IL-1β mediated BMSC (bone marrow mesenchymal stromal cell) senescence in mice after S. aureus infection. Importantly, IL-1β neutralizing antibody effectively alleviated BMSC senescence and bone loss caused by S. aureus infection in mice. In terms of molecular mechanism, we found IL-1β induced BMSC senescence by JNK/P53 and JNK/BCL2 pathways. Collectively, G-CSF promotes IL-1β production which induces BMSC senescence via JNK/P53 and JNK/BCL2 pathways, leading to S. aureus-induced bone loss. This study identified novel targets for preventing and treating S. aureus-induced bone loss in osteomyelitis.
Collapse
Affiliation(s)
- Mingrui Song
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingye Deng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ziyue Peng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fangfang Dai
- Huiqiao Medical Center, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yutian Wang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wen Shu
- Department of Trauma Orthopedics, Liuzhou People's Hospital, Liuzhou, Guangxi, China
| | - Xuyou Zhou
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinye Zhang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yilong Hou
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Zhou L, Li Y, Ma J, Zhang Q, Tang S, Zou K, Zeng Q, Huang H, Jin H, Zhang Q, Feng J. Role and mechanism of Actein on condylar bone metabolism in APOE deletion-induced osteoporotic mice. Bone 2024; 190:117304. [PMID: 39448001 DOI: 10.1016/j.bone.2024.117304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/18/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
AIMS To investigate the effects of Actein from Cimicifugae Rhizoma on condylar bone and cartilage in APOE deletion-induced osteoporotic mice, and to preliminarily explore the underlying mechanism. METHODS Sixty 8-week-old female mice were used, which underwent APOE-/- and ovariectomy procedures, followed by oral administration of Actein (15 mg/kg) and Atorvastatin Calcium (AC, 3 mg/kg) for eight weeks. Body weight, uterine weight, and systemic indexes related to bone metabolism and lipid metabolism were assessed in each group. Changes in condylar bone histomorphometric parameters were evaluated using Micro-CT. Morphological changes in the condyle were observed with Hematoxylin-Eosin (H&E), Safranin O/Fast Green, and Alcian Blue Hematoxylin/Orange G (ABH/OG) staining, with OARSI pathology scoring performed. Sirius red staining and immunofluorescence were used to determine the expression levels of Collagen I (Col I) and Collagen III (Col III) in bone matrix, and Col II in cartilage matrix. Immunohistochemistry assessed the relative expression levels of ALP and proteins associated with the Wnt/β-catenin/RUNX2 signaling pathway. RESULTS APOE-/- exacerbates ovariectomy -induced osteoporosis (OP) in condylar of mice. Actein and AC significantly reversed OP, improved bone mineral density (BMD), increased bone microarchitecture, and restored abnormal calcium and phosphorus metabolism in the blood and urine. Morphologically, APOE-/- and ovariectomy reduced condylar cartilage thickness, disrupted chondrocyte arrangement, chondrocyte cleavage, and clustered aggregation, resembling osteoarthritis (OA)-like changes. Actein and AC partially restored the disrupted chondrocyte arrangement, smoothed chondrocyte cleavage, and up-regulated the levels of chondrocyte matrix (Col II, aggrecan) and bone matrix (Col III, ALP). Actein reversed the OA process, potentially through the Wnt/β-catenin/RUNX2 signaling pathway. CONCLUSION APOE-/- and ovariectomy induced OP, leading to OA-like lesions in condylar of mice. Actein promoted cartilage repair and trabecular bone recovery by increasing extracellular matrix synthesis (Col II, Col III, aggrecan), reversing the OA process, possibly through the Wnt/β-catenin/RUNX2 signaling pathway.
Collapse
Affiliation(s)
- Linyi Zhou
- School/Hospital of Stomatology, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yuqian Li
- School/Hospital of Stomatology, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jinjin Ma
- Department of Stomatology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Qi Zhang
- Central Laboratories, Qingdao Municipal Hospital, Qingdao, Shandong 266071, China
| | - Shuhui Tang
- School/Hospital of Stomatology, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Kaiao Zou
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, China
| | - Qinghe Zeng
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, China
| | - Haipeng Huang
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, China
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310006, China
| | - Qiaoyan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Jianying Feng
- School/Hospital of Stomatology, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
4
|
Wang Y, Tong X, Shi X, Keswani T, Chatterjee E, Chen L, Li G, Lee K, Guo T, Yu Y. Chiral Cell Nanomechanics Originated in Clockwise/Counterclockwise Biofunctional Microarrays to Govern the Nuclear Mechanotransduction of Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2023; 15:48038-48049. [PMID: 37812566 DOI: 10.1021/acsami.3c11188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Cell chirality is extremely important for the evolution of cell morphogenesis to manipulate cell performance due to left-right asymmetry. Although chiral micro- and nanoscale biomaterials have been developed to regulate cell functions, how cell chirality affects cell nanomechanics to command nuclear mechanotransduction was ambiguous. In this study, chiral engineered microcircle arrays were prepared by photosensitive cross-linking synthesis on cell culture plates to control the clockwise/counterclockwise geometric topology of stem cells. Asymmetric focal adhesion and cytoskeleton structures could induce chiral cell nanomechanics measured by atomic force microscopy (AFM) nanoindentation in left-/right-handed stem cells. Cell nanomechanics could be enhanced when the construction of mature focal adhesion and the assembly of actin and myosin cytoskeletons were well organized in chiral engineered stem cells. Curvature angles had a negative effect on cell nanomechanics, while cell chirality did not change cytoskeletal mechanics. The biased cytoskeleton tension would engender different nuclear mechanotransductions by yes-associated protein (YAP) evaluation. The chiral stimuli were delivered into the nuclei to oversee nuclear behaviors. A strong cell modulus could activate high nuclear DNA synthesis activity by mechanotransduction. The results will bring the possibility of understanding the interplay of chiral cell nanomechanics and mechanotransduction in nanomedicines and biomaterials.
Collapse
Affiliation(s)
- Yongtao Wang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, 333 Nan Chen Road, Shanghai 200444, China
| | - Xiaolan Tong
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, 333 Nan Chen Road, Shanghai 200444, China
| | - Xiaohui Shi
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, 333 Nan Chen Road, Shanghai 200444, China
| | - Tarun Keswani
- Center for Immunological and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Emeli Chatterjee
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Lei Chen
- Department of Spine Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Spine Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Guoping Li
- Center for Immunological and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Kyubae Lee
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Tao Guo
- Department of Orthopaedics, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Yan Yu
- Department of Spine Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Spine Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| |
Collapse
|
5
|
Hu HY, Zhang ZZ, Jiang XY, Duan TH, Feng W, Wang XG. Hesperidin Anti-Osteoporosis by Regulating Estrogen Signaling Pathways. Molecules 2023; 28:6987. [PMID: 37836830 PMCID: PMC10574669 DOI: 10.3390/molecules28196987] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/27/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023] Open
Abstract
Osteoporosis (OP) is distinguished by a reduction in bone mass and degradation of bone micro-structure, frequently resulting in fractures. As the geriatric demographic expands, the incidence of affected individuals progressively rises, thereby exerting a significant impact on the quality of life experienced by individuals. The flavonoid compound hesperidin has been subject to investigation regarding its effects on skeletal health, albeit the precise mechanisms through which it operates remain ambiguous. This study utilized network pharmacology to predict the core targets and signaling pathways implicated in the anti-OP properties of hesperidin. Molecular docking and molecular dynamics simulations were employed to confirm the stability of the interaction between hesperidin and the core targets. The effects of hesperidin on osteoblastic cells MC3T3-E1 were assessed using MTT, ELISA, alkaline phosphatase assay, and RT-qPCR techniques. Furthermore, in vivo experiments were conducted to determine the potential protective effects of hesperidin on zebrafish bone formation and oxidative stress response. The results demonstrate that network pharmacology has identified 10 key target points, significantly enriched in the estrogen signaling pathway. Hesperidin exhibits notable promotion of MC3T3-E1 cell proliferation and significantly enhances ALP activity. ELISA measurements indicate an elevation in NO levels and a reduction in IL-6 and TNF-α. Moreover, RT-qPCR analysis consistently reveals that hesperidin significantly modulates the mRNA levels of ESR1, SRC, AKT1, and NOS3 in MC3T3-E1 cells. Hesperidin promotes osteogenesis and reduces oxidative stress in zebrafish. Additionally, we validate the stable and tight binding of hesperidin with ESR1, SRC, AKT1, and NOS3 through molecular dynamics simulations. In conclusion, our comprehensive analysis provides evidence that hesperidin may exert its effects on alleviating OP through the activation of the estrogen signaling pathway via ESR1. This activation leads to the upregulation of SRC, AKT, and eNOS, resulting in an increase in NO levels. Furthermore, hesperidin promotes osteoblast-mediated bone formation and inhibits pro-inflammatory cytokines, thereby alleviating oxidative stress associated with OP.
Collapse
Affiliation(s)
- Hong-Yao Hu
- Jilin Medical Products Administration, Changchun 130000, China;
| | - Ze-Zhao Zhang
- School of Pharmaceutical Sciences, Quality Evaluation & Standardization Hebei Province Engineering Research Center of Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050091, China; (Z.-Z.Z.); (X.-Y.J.); (T.-H.D.)
| | - Xiao-Ya Jiang
- School of Pharmaceutical Sciences, Quality Evaluation & Standardization Hebei Province Engineering Research Center of Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050091, China; (Z.-Z.Z.); (X.-Y.J.); (T.-H.D.)
| | - Tian-Hua Duan
- School of Pharmaceutical Sciences, Quality Evaluation & Standardization Hebei Province Engineering Research Center of Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050091, China; (Z.-Z.Z.); (X.-Y.J.); (T.-H.D.)
| | - Wei Feng
- School of Pharmaceutical Sciences, Quality Evaluation & Standardization Hebei Province Engineering Research Center of Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050091, China; (Z.-Z.Z.); (X.-Y.J.); (T.-H.D.)
| | - Xin-Guo Wang
- School of Pharmaceutical Sciences, Quality Evaluation & Standardization Hebei Province Engineering Research Center of Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050091, China; (Z.-Z.Z.); (X.-Y.J.); (T.-H.D.)
| |
Collapse
|
6
|
Hsu YH, Chen CN, Chang HI, Tsai HL, Chang YH, Cheng IS, Yang YS, Huang KY. Manipulation of osteogenic and adipogenic differentiation of human degenerative disc and ligamentum flavum derived progenitor cells using IL-1β, IL-19, and IL-20. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2023; 32:3413-3424. [PMID: 37563485 DOI: 10.1007/s00586-023-07878-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/22/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023]
Abstract
PURPOSE To elucidate whether pro-inflammatory cytokines might influence the commitment of intervertebral disc (IVD)- and ligamentum flavum (LF)-derived progenitor cells toward either osteogenesis or adipogenesis, specifically Interleukin-1β (IL-1β), IL-19, and IL-20. METHODS Sixty patients with degenerative spondylolisthesis and lumbar or lumbosacral spinal stenosis were included in the study. Injuries to the spine, infections, and benign or malignant tumors were excluded. From nine patient samples, IVD- and LF-derived cells were isolated after primary culture, and two clinical samples were excluded due to mycoplasma infection. The effects of IL-1β, IL-19, as well as IL-20 in regulating osteogenic and adipogenic differentiation in vitro were investigated. RESULTS Primary IVD- and LF-derived cells were found to have a similar cell morphology and profile of surface markers (CD44, CD90, and CD105) as placenta-derived mesenchymal stem cells (MSCs). Primary IVD/LF cells have a high capacity to differentiate into osteocytes and adipocytes. IL-19 had a tendency to promote adipogenesis. IL-20 inhibited osteogenesis and promoted adipogenesis; IL-1β promoted osteogenesis but inhibited adipogenesis. CONCLUSION IL-1β, IL-19, and IL-20 impact the adipogenic and osteogenic differentiation of IVD-derived and LF-derived cells. Modulating the expression of IL-1β, IL-19, and IL-20 provides a potential avenue for controlling cell differentiation of IVD- and LF-derived cells, which might have beneficial effect for degenerative spondylolisthesis and spinal stenosis.
Collapse
Affiliation(s)
- Yu-Hsiang Hsu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Nan Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi City, Taiwan
| | - Hsin-I Chang
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi City, Taiwan
| | - Hui-Ling Tsai
- Department of Orthopedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No. 138, Sheng-Li Road, Tainan, 704, Taiwan
| | - Yu-Hsien Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - I-Szu Cheng
- College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Shiuan Yang
- Education Center, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Kuo-Yuan Huang
- Department of Orthopedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No. 138, Sheng-Li Road, Tainan, 704, Taiwan.
| |
Collapse
|
7
|
Go YY, Lee CM, Chae SW, Song JJ. Regenerative capacity of trophoblast stem cell-derived extracellular vesicles on mesenchymal stem cells. Biomater Res 2023; 27:62. [PMID: 37370189 DOI: 10.1186/s40824-023-00396-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Human mesenchymal stem cells (MSCs) are therapeutic for clinical applications because of their excellent immunomodulatory and multiple lineage differentiation abilities at tissue injury sites. However, insufficient number of cells and lack of regenerative properties during in vitro expansion still limit the clinical applicability of MSC therapies. Here, we demonstrated a preconditioning strategy with trophoblast stem cell-derived extracellular vesicles (TSC-EVs) to boost the proliferation and regenerative capacity of MSCs. METHODS We employed cell proliferation analyses such as CCK8 and BrdU assays to determine the proliferation-promoting role of TSC-EVs on MSCs. Osteogenic effects of TSC-EVs on MSCs were assessed by alkaline phosphatase (ALP) activity, calcium assays, and calvarial bone defect animal models. For skin regenerative effects, skin wound mice model was exploited to analyze wound-healing rate in this study, as well as immunofluorescence and histological staining evaluates. We also performed the small RNA profiling and RNA-sequencing analyzes to understand the cellular mechanism of TSC-EVs on MSCs. RESULTS TSC-EVs significantly promoted MSC proliferation under xeno-free conditions and facilitated the therapeutic effects of MSCs, including osteogenesis, anti-senescence, and wound healing. Transcriptomic analysis also provided evidence that specific microRNAs in TSC-EVs and differentially expressed genes (DEGs) in TSC-EV-treated MSCs showed the possibility of TSC-EVs triggering the regenerative abilities of MSCs with cytokine interaction. Hence, we found that NGF/Akt signaling mediated the regenerative effects of TSC-EVs on MSCs as a particular cellular signaling pathway. CONCLUSION The results of this study demonstrated the functional properties of TSC-EVs on MSCs for MSC-based therapeutic applications, suggesting that TSC-EVs may serve as a potential preconditioning source for MSC therapy in the clinical field of regenerative medicine.
Collapse
Affiliation(s)
- Yoon-Young Go
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, 08308, Republic of Korea
| | - Chan-Mi Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea
| | - Sung-Won Chae
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea.
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, 08308, Republic of Korea.
| |
Collapse
|
8
|
Jing Y, Zhou J, Guo F, Yu L, Ren X, Yin X. Betaine regulates adipogenic and osteogenic differentiation of hAD-MSCs. Mol Biol Rep 2023; 50:5081-5089. [PMID: 37101008 DOI: 10.1007/s11033-023-08404-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/23/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND With an ageing population, the incidence of bone loss and obesity are increasing. Numerous studies emphasized the multidirectional differentiation ability of mesenchymal stem cells (MSCs), and reported betaine modulated the osteogenic differentiation and adipogenic differentiation of MSCs in vitro. We wondered how betaine affected the differentiation of hAD-MSCs and hUC-MSCs. METHODS AND RESULTS ALP staining and alizarin red S (ARS) staining were proved 10 mM betaine significantly increased the number of ALP-positive cells and plaque calcified extracellular matrices, accompanying by the up-regulation of OPN, Runx-2 and OCN. Oil red O staining demonstrated the number and size of lipid droplets were reduced, the expression of adipogenic master genes such as PPARγ, CEBPα and FASN were down-regulated simultaneously. For further investigating the mechanism of betaine on hAD-MSCs, RNA-seq was performed in none-differentiation medium. The Gene Ontology (GO) analysis showed fat cell differentiation and bone mineralization function terms were enriched, and KEGG showed PI3K-Akt signaling pathway, cytokine-cytokine receptor interaction and ECM-receptor interaction pathways were enriched in betaine treated hAD-MSCs, demonstrated betaine had a positive inducing effect on osteogenic of hAD-MSCs in the non-differentiation medium in vitro, which is opposite to the effect on adipogenic differentiation. CONCLUSIONS Our study demonstrated that betaine promoted osteogenic and compromised adipogenic differentiation of hUC-MSCs and hAD-MSCs upon low concentration administration. PI3K-Akt signaling pathway, cytokine-cytokine receptor interaction and ECM-receptor interaction were significantly enriched under betaine-treated. We showed hAD-MSCs were more sensitive to betaine stimulation and have a better differentiation ability than hUC-MSCs. Our results contributed to the exploration of betaine as an aiding agent for MSCs therapy.
Collapse
Affiliation(s)
- Yue Jing
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, Liaoning Province, China
| | - Jian Zhou
- College of Pharmaceutical Sciences, Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Fenghua Guo
- Jiangsu Pulu Rui Medical Technology Co., Ltd, Xuzhou, Jiangsu Province, China
| | - Lin Yu
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, Liaoning Province, China
| | - Xiaomeng Ren
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, Liaoning Province, China
| | - Xiushan Yin
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, Liaoning Province, China.
| |
Collapse
|
9
|
Entz L, Falgayrac G, Chauveau C, Pasquier G, Lucas S. The extracellular matrix of human bone marrow adipocytes and glucose concentration differentially alter mineralization quality without impairing osteoblastogenesis. Bone Rep 2022; 17:101622. [PMID: 36187598 PMCID: PMC9519944 DOI: 10.1016/j.bonr.2022.101622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/09/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022] Open
Abstract
Bone marrow adipocytes (BMAds) accrue in various states of osteoporosis and interfere with bone remodeling through the secretion of various factors. However, involvement of the extracellular matrix (ECM) produced by BMAds in the impairment of bone marrow mesenchymal stromal cell (BM-MSC) osteoblastogenesis has received little attention. In type 2 diabetes (T2D), skeletal fragility is associated with several changes in bone quality that are incompletely understood, and BMAd quantity increases in relationship to poor glycemic control. Considering their altered phenotype in this pathophysiological context, we aimed to determine the contribution of the ECM of mature BMAds to osteoblastogenesis and mineralization quality in the context of chronic hyperglycemia. Human BM-MSCs were differentiated for 21 days in adipogenic medium containing either a normoglycemic (LG, 5.5 mM) or a high glucose concentration (HG, 25 mM). The ECM laid down by BMAds were devitalized through cell removal to examine their impact on the proliferation and differentiation of BM-MSCs toward osteoblastogenesis in LG and HG conditions. Compared to control plates, both adipocyte ECMs promoted cell adhesion and proliferation. As shown by the unmodified RUNX2 and osteocalcin mRNA levels, BM-MSC commitment in osteoblastogenesis was hampered by neither the hyperglycemic condition nor the adipocyte matrices. However, adipocyte ECMs or HG condition altered the mineralization phase with perturbed expression levels of type 1 collagen, MGP and osteopontin. Despite higher ALP activity, mineralization levels per cell were decreased for osteoblasts grown on adipocyte ECMs compared to controls. Raman spectrometry revealed that culturing on adipocyte matrices specifically prevents type-B carbonate substitution and favors collagen crosslinking, in contrast to exposure to HG concentration alone. Moreover, the mineral to organic ratio was disrupted according to the presence of adipocyte ECM and the glucose concentration used for adipocyte or osteoblast culture. HG concentration and adipocyte ECM lead to different defects in mineralization quality, recapitulating contradictory changes reported in T2D osteoporosis. Our study shows that ECMs from BMAds do not impair osteoblastogenesis but alter both the quantity and quality of mineralization partly in a glucose concentration-dependent manner. This finding sheds light on the involvement of BMAds, which should be considered in the compromised bone quality of T2D and osteoporosis patients more generally. Glucose level alters the Extracellular Matrix composition of Bone Marrow adipocytes. Osteoblastogenesis on adipocyte ECMs is unaltered but produced less mineral amount. The quality of the mineral is altered differently by adipocyte ECMs or glucose levels. The presence of BM adipocytes should be valued in damaged osteoporosis bone quality.
Collapse
Key Words
- AGEs, Advanced glycation end-products
- BM-MSC, Bone marrow mesenchymal stromal cell
- BMAd, Bone marrow adipocyte
- ECM, Extracellular matrix
- ECMBMAd HG, Extracellular matrix obtained from BMAds cultured in HG concentration
- ECMBMAd LG, Extracellular matrix obtained from BMAds cultured in LG concentration
- ECMBMAd, Extracellular matrix obtained from BMAds
- Extracellular matrix
- GAG, glycosaminoglycan
- HA, hydroxyapatite
- HG, High glucose
- Hyperglycemia
- LG, Low glucose
- LGM, Low glucose and mannitol
- Marrow adipocytes
- Osteoblast
- Osteoporosis
- Skeletal mesenchymal stromal cells
- T2D, Type 2 diabetes
Collapse
|
10
|
Chan NT, Lee MS, Wang Y, Galipeau J, Li WJ, Xu W. CTR9 drives osteochondral lineage differentiation of human mesenchymal stem cells via epigenetic regulation of BMP-2 signaling. SCIENCE ADVANCES 2022; 8:eadc9222. [PMID: 36383652 PMCID: PMC9668309 DOI: 10.1126/sciadv.adc9222] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/19/2022] [Indexed: 05/06/2023]
Abstract
Cell fate determination of human mesenchymal stem/stromal cells (hMSCs) is precisely regulated by lineage-specific transcription factors and epigenetic enzymes. We found that CTR9, a key scaffold subunit of polymerase-associated factor complex (PAFc), selectively regulates hMSC differentiation to osteoblasts and chondrocytes, but not to adipocytes. An in vivo ectopic osteogenesis assay confirmed the essentiality of CTR9 in hMSC-derived bone formation. CTR9 counteracts the activity of Enhancer Of Zeste 2 (EZH2), the epigenetic enzyme that deposits H3K27me3, in hMSCs. Accordingly, CTR9 knockdown (KD) hMSCs gain H3K27me3 mark, and the osteogenic differentiation defects of CTR9 KD hMSCs can be partially rescued by treatment with EZH2 inhibitors. Transcriptome analyses identified bone morphology protein-2 (BMP-2) as a downstream effector of CTR9. BMP-2 secretion, membrane anchorage, and the BMP-SMAD pathway were impaired in CTR9 KD MSCs, and the effects were rescued by BMP-2 supplementation. This study uncovers an epigenetic mechanism engaging the CTR9-H3K27me3-BMP-2 axis to regulate the osteochondral lineage differentiation of hMSCs.
Collapse
Affiliation(s)
- Ngai Ting Chan
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin Carbone Comprehensive Cancer Center, Madison, WI 53706, USA
| | - Ming-Song Lee
- Department of Orthopedics and Rehabilitation, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yidan Wang
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin Carbone Comprehensive Cancer Center, Madison, WI 53706, USA
| | - Jacques Galipeau
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wan-Ju Li
- Department of Orthopedics and Rehabilitation, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, Wisconsin Institute for Medical Research, University of Wisconsin Carbone Comprehensive Cancer Center, Madison, WI 53706, USA
| |
Collapse
|
11
|
Morphological Dependence of Breast Cancer Cell Responses to Doxorubicin on Micropatterned Surfaces. Polymers (Basel) 2022; 14:polym14142761. [PMID: 35890536 PMCID: PMC9323815 DOI: 10.3390/polym14142761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022] Open
Abstract
Cell morphology has been widely investigated for its influence on the functions of normal cells. However, the influence of cell morphology on cancer cell resistance to anti-cancer drugs remains unclear. In this study, micropatterned surfaces were prepared and used to control the spreading area and elongation of human breast cancer cell line. The influences of cell adhesion area and elongation on resistance to doxorubicin were investigated. The percentage of apoptotic breast cancer cells decreased with cell spreading area, while did not change with cell elongation. Large breast cancer cells had higher resistance to doxorubicin, better assembled actin filaments, higher DNA synthesis activity and higher expression of P-glycoprotein than small breast cancer cells. The results suggested that the morphology of breast cancer cells could affect their resistance to doxorubicin. The influence was correlated with cytoskeletal organization, DNA synthesis activity and P-glycoprotein expression.
Collapse
|
12
|
Yang Y, Liu S, He C, Chen Z, Lyu T, Zeng L, Wang L, Zhang F, Chen H, Zhao RC. Long Non-coding RNA Regulation of Mesenchymal Stem Cell Homeostasis and Differentiation: Advances, Challenges, and Perspectives. Front Cell Dev Biol 2021; 9:711005. [PMID: 34368161 PMCID: PMC8339964 DOI: 10.3389/fcell.2021.711005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/21/2021] [Indexed: 12/25/2022] Open
Abstract
Given the self-renewal, multi-differentiation, immunoregulatory, and tissue maintenance properties, mesenchymal stem cells (MSCs) are promising candidates for stem cell-based therapies. Breakthroughs have been made in uncovering MSCs as key contributors to homeostasis and the regenerative repair of tissues and organs derived from three germ layers. MSC differentiation into specialized cell types is sophisticatedly regulated, and accumulating evidence suggests long non-coding RNAs (lncRNAs) as the master regulators of various biological processes including the maintenance of homeostasis and multi-differentiation functions through epigenetic, transcriptional, and post-translational mechanisms. LncRNAs are ubiquitous and generally referred to as non-coding transcripts longer than 200 bp. Most lncRNAs are evolutionary conserved and species-specific; however, the weak conservation of their sequences across species does not affect their diverse biological functions. Although numerous lncRNAs have been annotated and studied, they are nevertheless only the tip of the iceberg; the rest remain to be discovered. In this review, we characterize MSC functions in homeostasis and highlight recent advances on the functions and mechanisms of lncRNAs in regulating MSC homeostasis and differentiation. We also discuss the current challenges and perspectives for understanding the roles of lncRNAs in MSC functions in homeostasis, which could help develop promising targets for MSC-based therapies.
Collapse
Affiliation(s)
- Yanlei Yang
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Beijing Key Laboratory (No. BZO381), School of Basic Medicine, Center of Excellence in Tissue Engineering, Peking Union Medical College Hospital, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Suying Liu
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Chengmei He
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhilei Chen
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Taibiao Lyu
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Liuting Zeng
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Li Wang
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Fengchun Zhang
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hua Chen
- Key Laboratory of the Ministry of Education, Department of Rheumatology and Clinical Immunology, Clinical Immunology Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Robert Chunhua Zhao
- Beijing Key Laboratory (No. BZO381), School of Basic Medicine, Center of Excellence in Tissue Engineering, Peking Union Medical College Hospital, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
13
|
Transcriptome Analysis of Egg Yolk Sialoglycoprotein on Osteogenic Activity in MC3T3-E1 Cells. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11146428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this study, the effects of egg yolk sialoglycoprotein (EYG) on osteogenesis in MC3T3-E1 cells were investigated and the DEGs (differentially expressed genes) were explored by transcriptome analysis. The results found that EYG effectively increased cell proliferation, enhanced ALP activity, promoted the secretion of extracellular matrix protein COL-I and OCN, enhanced bone mineralization activity, exhibiting good osteogenic activity. Further study of the mechanism was explored through transcriptome analysis. Transcriptome analysis showed that 123 DEGs were triggered by EYG, of which 78 genes were downregulated and 45 genes were upregulated. GO (gene ontology) analysis showed that EYG mainly caused differences in gene expression of biological processes and cell composition categories in the top 30 most enriched items. KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis showed that EYG inhibited inflammatory factors and downregulated inflammation-related pathways. The results also showed EYG regulated such genes as COL2A1, COL4A1 and COL4A2 to up-regulate pathways including ECM–receptor interaction, focal adhesion and protein digestion and absorption, enhancing the proliferation and differentiation of osteoblasts. Gene expression of COL-I, Runx2, BMP2 and β-catenin was determined by qRT-PCR for verification, which found that EYG significantly increased COL-I, Runx2, BMP2 and β-catenin gene expression, suggesting that BMP-2 mediated osteogenesis pathway was activated.
Collapse
|
14
|
Antich C, Jiménez G, de Vicente J, López‐Ruiz E, Chocarro‐Wrona C, Griñán‐Lisón C, Carrillo E, Montañez E, Marchal JA. Development of a Biomimetic Hydrogel Based on Predifferentiated Mesenchymal Stem-Cell-Derived ECM for Cartilage Tissue Engineering. Adv Healthc Mater 2021; 10:e2001847. [PMID: 33646595 PMCID: PMC11468687 DOI: 10.1002/adhm.202001847] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/06/2021] [Indexed: 12/20/2022]
Abstract
The use of decellularized extracellular matrix (dECM) as a biomaterial has been an important step forward for the development of functional tissue constructs. In addition to tissues and organs, cell cultures are gaining a lot of attention as an alternative source of dECM. In this work, a novel biomimetic hydrogel is developed based on dECM obtained from mesenchymal stem cells (mdECM) for cartilage tissue engineering. To this end, cells are seeded under specific culture conditions to generate an early chondrogenic extracellular matrix (ECM) providing cues and elements necessary for cartilage development. The composition is determined by quantitative, histological, and mass spectrometry techniques. Moreover, the decellularization process is evaluated by measuring the DNA content and compositional analyses, and the hydrogel is formulated at different concentrations (3% and 6% w/v). Results show that mdECM derived hydrogels possess excellent biocompatibility and suitable physicochemical and mechanical properties for their injectability. Furthermore, it is evidenced that this hydrogel is able to induce chondrogenesis of mesenchymal stem cells (MSCs) without supplemental factors and, furthermore, to form hyaline cartilage-like tissue after in vivo implantation. These findings demonstrate for the first time the potential of this hydrogel based on mdECM for applications in cartilage repair and regeneration.
Collapse
Affiliation(s)
- Cristina Antich
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranada18016Spain
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
| | - Gema Jiménez
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
- Department of Health ScienceFaculty of Experimental ScienceUniversity of JaénJaén23071Spain
| | - Juan de Vicente
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
- Biocolloid and Fluid Physics GroupDepartment of Applied PhysicsFaculty of SciencesUniversity of GranadaGranada18071Spain
| | - Elena López‐Ruiz
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
- Department of Health ScienceFaculty of Experimental ScienceUniversity of JaénJaén23071Spain
| | - Carlos Chocarro‐Wrona
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranada18016Spain
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
| | - Carmen Griñán‐Lisón
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranada18016Spain
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
| | - Esmeralda Carrillo
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranada18016Spain
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
| | - Elvira Montañez
- Department of Orthopedic Surgery and TraumatologyVirgen de la Victoria University HospitalMálaga29010Spain
- Biomedical Research Institute of Malaga (IBIMA)Virgen de la Victoria University HospitalMálaga29010Spain
| | - Juan A. Marchal
- Department of Human Anatomy and EmbryologyFaculty of MedicineUniversity of GranadaGranada18016Spain
- Instituto de Investigación Biosanitaria ibs. GRANADAUniversity of GranadaGranada18014Spain
- Biopathology and Regenerative Medicine Institute (IBIMER)Centre for Biomedical ResearchUniversity of GranadaGranada18100Spain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranada18016Spain
| |
Collapse
|
15
|
Chen Y, Lee K, Kawazoe N, Yang Y, Chen G. ECM scaffolds mimicking extracellular matrices of endochondral ossification for the regulation of mesenchymal stem cell differentiation. Acta Biomater 2020; 114:158-169. [PMID: 32738504 DOI: 10.1016/j.actbio.2020.07.049] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/02/2020] [Accepted: 07/26/2020] [Indexed: 12/11/2022]
Abstract
Endochondral ossification (ECO) is an important process of bone tissue development. During ECO, extracellular matrices (ECMs) are essential factors to control cell functions and induce bone regeneration. However, the exact role of ECO ECMs on stem cell differentiation remains elusive. In this study, ECM scaffolds were prepared to mimic the ECO-related ECM microenvironments and their effects on stem cell differentiation were compared. Four types of ECM scaffolds mimicking the ECMs of stem cells (SC), chondrogenic (CH), hypertrophic (HY) and osteogenic (OS) stages were prepared by controlling differentiation of human bone marrow-derived mesenchymal stem cells (MSCs) at different stages. Composition of the ECM scaffolds was dependent on the differentiation stage of MSCs. They showed different influence on osteogenic differentiation of MSCs. HY ECM scaffold had the most promotive effect on osteogenic differentiation of MSCs. CH ECM and OS ECM scaffolds showed moderate effect, while SC ECM scaffold had the lowest effect on osteogenic differentiation of MSCs. Their effects on chondrogenic or adipogenic differentiation were not significantly different. The results suggested that the engineered HY ECM scaffold had superior effect for osteogenic differentiation of MSCs. Statement of significance ECM scaffolds mimicking endochondral ossification-related ECM microenvironments are pivotal for elucidation of their roles in regulation of stem cell functions and bone tissue regeneration. This study offers a method to prepare ECM scaffolds that mimic the ECMs from cells at hypertrophic, osteogenic, chondrogenic and stem cell stages. Their composition and impacts on osteogenic differentiation of MSCs were compared. The hypertrophic ECM scaffold had the highest promotive effect on osteogenic differentiation of MSCs. The results advance our understanding about the role of ECO ECMs in regulation of stem cell functions and provide perspective for bone defect repair strategies.
Collapse
|
16
|
Hu X, Liu X, Lv L, Zhang X, Liu Y, Zhang P, Zhou Y. UNC-5 netrin receptor B regulates adipogenesis of human adipose-derived stem cells through JNK pathway. J Oral Rehabil 2020; 47 Suppl 1:91-98. [PMID: 32762046 DOI: 10.1111/joor.13067] [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: 12/11/2018] [Revised: 07/09/2020] [Accepted: 07/29/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND There is a balance between adipogenic differentiation and osteogenic differentiation of human adipose-derived stem cells (hASCs). It is essential to explore the mechanism of hASCs lineage commitment. In our previous study, UNC-5 netrin receptor B (UNC5B) was identified as a positive regulator for osteogenesis. OBJECTIVE To further explore the potential roles and mechanisms of UNC5B during adipogenic differentiation and to provide a new method to regulate adipogenesis and osteogenesis of hASCs. METHODS Lentivirus containing UNC5B shRNA was used for UNC5B knockdown. Plasmids overexpressing UNC5B gene were used for UNC5B upregulation. To investigate the role of UNC5B in adipogenesis in vitro and in vivo, Oil Red O staining, RT-qPCR and transplantation into nude mice were performed. Western blotting analyses were performed to explore the mechanisms of UNC5B in adipogenic differentiation. RESULTS UNC5B expression in hASCs was significantly increased during adipogenic differentiation. Knockdown of UNC5B enhanced adipogenic differentiation in vitro. Both H&E staining and Oil Red O staining showed more adipose tissue-like constructs in UNC5B-knockdown cells in vivo. Upregulation of UNC5B significantly impaired adipogenic differentiation in vitro. Downregulation of UNC5B could increase phosphorylation of JNK in hASCs. JNK inhibitors reduced adipogenic differentiation of hASCs. CONCLUSION Our findings showed that UNC5B inhibited adipogenesis of hASCs through JNK signalling. As a whole, UNC5B regulates both adipogenesis and osteogenesis of hASCs.
Collapse
Affiliation(s)
- Xinyi Hu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Department of Stomatology, Shenzhen University General Hospital, Xili University Town, Shenzhen, China
| | - Xuejiao Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China
| | - Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
17
|
Chen Y, Lee K, Yang Y, Kawazoe N, Chen G. PLGA-collagen-ECM hybrid meshes mimicking stepwise osteogenesis and their influence on the osteogenic differentiation of hMSCs. Biofabrication 2020; 12:025027. [DOI: 10.1088/1758-5090/ab782b] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
18
|
Yan D, Tang B, Yan L, Zhang L, Miao M, Chen X, Sui G, Zhang Q, Liu D, Wang H. Sodium Selenite Improves The Therapeutic Effect Of BMSCs Via Promoting The Proliferation And Differentiation, Thereby Promoting The Hematopoietic Factors. Onco Targets Ther 2020; 12:9685-9696. [PMID: 32009801 PMCID: PMC6859959 DOI: 10.2147/ott.s209937] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
Purpose Sodium selenite (Na2SeO3) has been known to restore the antioxidant capacity of bone marrow mesenchymal stem cells (BMSCs), reduce the production of reactive oxygen species (ROS) in the cells, and promote cell proliferation and inhibit cell apoptosis. However, it is still not clear whether selenium can mediate the differentiation and inhibit the induced hemagglutination of BMSCs. In this study, we attempted to explore the effect of Na2SeO3 on these aspects of BMSCs. Methods We evaluated the fate of the MSCs isolated from the bone marrow of mice by studying their differentiation and proliferation after treatment with Na2SeO3. We also simultaneously evaluated the coagulation reaction induced by Na2SeO3-treated BMSCs in vitro. Results While the mice-derived BMSCs expressed CD44, CD73, CD90, and CD105, they did not express CD45. The morphology of the derived cells was homogeneously elongated. These results showed that the isolated cells are indeed BMSCs. We found that 0.1 μM and 1 μM of Na2SeO3 promoted the proliferation and apoptosis of BMSCs, respectively. This showed that Na2SeO3 can be toxic and exert certain side effects on the BMSCs. The results of the osteogenic and adipogenic assay showed that 0.1 μM Na2SeO3 could significantly promote the osteogenic and adipogenic differentiation of BMSCs by upregulating the lipid factors (LPL and PPRAG) and osteogenic factors, RUNX2, COL1, and BGP, in a concentration-dependent manner. Coagulation experiments in animals (mice and rats) revealed that Na2SeO3 can reduce the coagulation time of BMSCs in a concentration-dependent manner, which is related to the high expression of hematopoietic factors (SDF-1α, GM-CSF, IL-7, IL-8, IL-11, and SCF). Conclusion Na2SeO3 promotes the proliferation and differentiation as well as reduces the coagulation time of BMSCs, and this effect might enhance the therapeutic effect of BMSCs.
Collapse
Affiliation(s)
- Dongmei Yan
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Botao Tang
- Department of Cardiology, Heilongjiang Red Cross Hospital, Harbin, People's Republic of China
| | - Lixin Yan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Lei Zhang
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Meijuan Miao
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Xi Chen
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Guangyi Sui
- Ethics Committee, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, People's Republic of China
| | - Qi Zhang
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Daoyuan Liu
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Hui Wang
- Department of Blood Transfusion, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|
19
|
Hoshiba T, Yokoyama N. Decellularized extracellular matrices derived from cultured cells at stepwise myogenic stages for the regulation of myotube formation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118658. [PMID: 31978502 DOI: 10.1016/j.bbamcr.2020.118658] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 11/30/2022]
Abstract
The regulation of stem cell differentiation is key for muscle tissue engineering and regenerative medicine. To this end, various substrates mimicking the native extracellular matrix (ECM) have been developed with consideration of the mechanical, topological, and biochemical properties. However, mimicking the biochemical properties of the native ECM is difficult due to its compositional complexity. To develop substrates that mimic the native ECM and its biochemical properties, decellularization is typically used. Here, substrates mimicking the native ECM at each myogenic stage are prepared as stepwise myogenesis-mimicking matrices via the in vitro myogenic culture of C2C12 myoblasts and decellularization. Cells adhered to the stepwise myogenesis-mimicking matrices at similar levels. However, the matrices derived from cells at the myogenic early stage suppressed cell growth and promoted myogenesis. This promotion of myogenesis was potentially due to the suppression of the activation of endogenous BMP signaling following the suppression of the expression of the myogenic-inhibitory factors, Id2 and Id3. Our stepwise myogenesis-mimicking matrices will be suitable ECM models for basic biological research and myogenesis of stem cells. Further, these matrices will provide insights that improve the efficacy of decellularized ECM for muscle repair.
Collapse
Affiliation(s)
- Takashi Hoshiba
- Biotechnology Group, Tokyo Metropolitan Industrial Technology Research Institute, 2-4-10 Aomi, Koto-ku, Tokyo 135-0064, Japan; Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.
| | - Natsumi Yokoyama
- Yamagata Prefectural Yonezawa Kojokan Senior High School, 1101 Oh-aza, Sasano, Yonezawa, Yamagata 992-1443, Japan
| |
Collapse
|
20
|
Decellularized hASCs-derived matrices as biomaterials for 3D in vitro approaches. Methods Cell Biol 2020; 156:45-58. [DOI: 10.1016/bs.mcb.2019.11.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
21
|
Benayahu D, Wiesenfeld Y, Sapir-Koren R. How is mechanobiology involved in mesenchymal stem cell differentiation toward the osteoblastic or adipogenic fate? J Cell Physiol 2019; 234:12133-12141. [PMID: 30633367 DOI: 10.1002/jcp.28099] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 12/07/2018] [Indexed: 12/28/2022]
Abstract
Mechanobiology plays a major role in transducing physical cues from the dynamic cellular environment into biochemical modifications that promote cell-specific differentiation paths. Mesenchymal stem cells in the bone marrow or in other mesenchymal tissues will differentiate according to the expression of transcription factors (TFs) that govern their lineage commitment. The favoring of either osteogenic or adipogenic differentiation relies on TF expression as well as mechanical properties of the cells' niche that are translated into the activation of certain signaling pathways. Physical factors can induce significant shifts in bipotential lineage commitment between osteogenesis and adipogenesis. The stiffness of the extracellular matrix (ECM) surrounding a cell, varying greatly from rigid environments close to the bone surface to softer regions in the bone marrow, can influence the path of differentiation. Additionally, mechanical loading through exercise appears to favor osteogenesis whereas disuse conditions seem to promote adipogenesis.
Collapse
Affiliation(s)
- Dafna Benayahu
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yarden Wiesenfeld
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rony Sapir-Koren
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
22
|
Moeinzadeh S, Monavarian M, Kader S, Jabbari E. Sequential Zonal Chondrogenic Differentiation of Mesenchymal Stem Cells in Cartilage Matrices. Tissue Eng Part A 2018; 25:234-247. [PMID: 30146939 DOI: 10.1089/ten.tea.2018.0083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
IMPACT STATEMENT The higher regenerative capacity of fetal articular cartilage compared with the adult is rooted in differences in cell density and matrix composition. We hypothesized that the zonal organization of articular cartilage can be engineered by encapsulation of mesenchymal stem cells in a single superficial zone-like matrix followed by sequential addition of zone-specific growth factors within the matrix, similar to the process of fetal cartilage development. The results demonstrate that the zonal organization of articular cartilage can potentially be regenerated using an injectable, monolayer cell-laden hydrogel with sequential release of growth factors.
Collapse
Affiliation(s)
- Seyedsina Moeinzadeh
- 1 Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina
| | - Mehri Monavarian
- 1 Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina
| | - Safaa Kader
- 1 Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina.,2 Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina
| | - Esmaiel Jabbari
- 1 Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
23
|
Baumgartner W, Otto L, Hess SC, Stark WJ, Märsmann S, Bürgisser GM, Calcagni M, Cinelli P, Buschmann J. Cartilage/bone interface fabricated under perfusion: Spatially organized commitment of adipose‐derived stem cells without medium supplementation. J Biomed Mater Res B Appl Biomater 2018; 107:1833-1843. [DOI: 10.1002/jbm.b.34276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/27/2018] [Accepted: 10/17/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Walter Baumgartner
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Lukas Otto
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Samuel C. Hess
- Institute for Chemical‐ and BioengineeringDepartment of Chemistry and Applied Biosciences ETH Zürich, Zürich Switzerland
| | - Wendelin J. Stark
- Institute for Chemical‐ and BioengineeringDepartment of Chemistry and Applied Biosciences ETH Zürich, Zürich Switzerland
| | - Sonja Märsmann
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
- Division of Trauma SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | | | - Maurizio Calcagni
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Paolo Cinelli
- Division of Trauma SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Johanna Buschmann
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| |
Collapse
|
24
|
Liu LL, Liu L, Liu HH, Ren SS, Dou CY, Cheng PP, Wang CL, Wang LN, Chen XL, Zhang H, Chen MT. Levamisole suppresses adipogenesis of aplastic anaemia-derived bone marrow mesenchymal stem cells through ZFP36L1-PPARGC1B axis. J Cell Mol Med 2018; 22:4496-4506. [PMID: 29993187 PMCID: PMC6111807 DOI: 10.1111/jcmm.13761] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/08/2018] [Indexed: 11/26/2022] Open
Abstract
Aplastic anaemia (AA) is a life‐threatening hematopoietic disorder characterized by hypoplasia and pancytopenia with increasing fat cells in the bone marrow (BM). The BM‐derived mesenchymal stem cells (MSCs) from AA are more susceptible to be induced into adipogenic differentiation compared with that from control, which may be causatively associated with the fatty BM and defective hematopoiesis of AA. Here in this study, we first demonstrated that levamisole displayed a significant suppressive effect on the in vitro adipogenic differentiation of AA BM‐MSCs. Mechanistic investigation revealed that levamisole could increase the expression of ZFP36L1 which was subsequently demonstrated to function as a negative regulator of adipogenic differentiation of AA BM‐MSCs through lentivirus‐mediated ZFP36L1 knock‐down and overexpression assay. Peroxisome proliferator‐activated receptor gamma coactivator 1 beta (PPARGC1B) whose 3′‐untranslated region bears adenine‐uridine‐rich elements was verified as a direct downstream target of ZFP36L1, and knock‐down of PPARGC1B impaired the adipogenesis of AA BM‐MSCs. Collectively, our work demonstrated that ZFP36L1‐mediated post‐transcriptional control of PPARGC1B expression underlies the suppressive effect of levamisole on the adipogenic differentiation of AA BM‐MSCs, which not only provides novel therapeutic targets for alleviating the BM fatty phenomenon of AA patients, but also lays the theoretical and experimental foundation for the clinical application of levamisole in AA therapy.
Collapse
Affiliation(s)
- Lu-Lu Liu
- Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, China
| | - Lei Liu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Hai-Hui Liu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, China.,Department of Graduate School, Jining Medical University, Jining, China
| | - Sai-Sai Ren
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Cui-Yun Dou
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Pan-Pan Cheng
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Cui-Ling Wang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Li-Na Wang
- Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, China
| | - Xiao-Li Chen
- Department of Graduate School, Jining Medical University, Jining, China
| | - Hao Zhang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Ming-Tai Chen
- Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, China
| |
Collapse
|
25
|
Baxter-Holland M, Dass CR. Pigment epithelium-derived factor: a key mediator in bone homeostasis and potential for bone regenerative therapy. J Pharm Pharmacol 2018; 70:1127-1138. [DOI: 10.1111/jphp.12942] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 05/19/2018] [Indexed: 01/02/2023]
Abstract
Abstract
Objectives
Pigment epithelium-derived factor (PEDF), a multifunctional endogenous glycoprotein, has a very wide range of biological actions, notably in bone homeostasis. The question has been raised regarding the place of PEDF in the treatment of bone disorders and osteosarcoma, and its potential for tumour growth suppression.
Methods
The PubMed database was used to compile this review.
Key findings
Pigment epithelium-derived factor's actions in osteoid tissues include promoting mesenchymal stem cell commitment to osteoblasts, increasing matrix mineralisation, and promoting osteoblast proliferation. It shows potential to improve therapeutic outcomes in treatment of multiple cancer types and regrowth of bone after trauma or resection in animal studies. PEDF may possibly have a reduced adverse effect profile compared with current osteo-regenerative treatments; however, there is currently very limited evidence regarding the safety or efficacy in human models.
Summary
Pigment epithelium-derived factor is very active within the body, particularly in osseous tissue, and its physiological actions give it potential for treatment of both bone disorders and multiple tumour types. Further research is needed to ascertain the adverse effects and safety profile of PEDF as a therapeutic agent.
Collapse
Affiliation(s)
- Mia Baxter-Holland
- School of Pharmacy and Biomedical Science, Curtin University, Perth, WA, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Perth, WA, Australia
- Curtin Health Innovation Research Institute, Bentley, WA, Australia
| |
Collapse
|
26
|
Chang SF, Yeh CC, Chen PJ, Chang HI. The Impact of Lipid Types and Liposomal Formulations on Osteoblast Adiposity and Mineralization. Molecules 2018; 23:molecules23010095. [PMID: 29301300 PMCID: PMC6017718 DOI: 10.3390/molecules23010095] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/22/2017] [Accepted: 12/29/2017] [Indexed: 12/22/2022] Open
Abstract
Recent studies have demonstrated that fat accumulation in bone cells is detrimental to bone mass. Both adipocytes and osteoblasts are derived from common multipotent mesenchymal stem cells (MSCs) and hence the presence of fat may increase adipocyte proliferation, differentiation and fat accumulation while inhibiting osteoblast differentiation and bone formation. Lipids are common constituents in supramolecular vesicles (e.g., micelles or liposomes) that serve as drug delivery systems. Liposomal formulations such as Meriva® were proven to decrease joint pain and improve joint function in osteoarthritis (OA) patients. In this study, we evaluated how lipid types and liposomal formulations affect osteoblast behavior including cell viability, differentiation, mineralization and inflammation. Various liposomal formulations were prepared using different types of lipids, including phosphatidylcholine (PC), 1,2-dioleoyl-sn-glycero-3-phospho-ethanolamine (DOPE), cholesterol (Chol), 3β-[N-(N′,N′-dimethylaminoethane)-carbamoyl] cholesterol hydrochloride (DC-cholesterol HCl), and 1,2-dioleoyl-3-trimethylammonium-propane chloride salt (DOTAP) to investigate the impact on osteoblast differentiation and inflammation. The results indicated that cationic lipids, DC-cholesterol and DOTAP, presented higher dose-dependent cytotoxicity and caused high level of inflammatory responses. Due to the natural properties of lipids, all the lipids can induce lipid droplet formation in osteoblasts but the level of lipid droplet accumulation was different. In comparison with cationic lipids, neutral lipids induced less adiposity, and maintained high osteoblast mineralization. Similar to previous researches, we also confirmed an inverse relationship between lipid droplet formation and osteoblast mineralization in 7F2 mouse osteoblasts. Importantly, PC containing liposomes (PC only and PC/DOTAP) suppressed IL-1β-induced gene expression of COX-2 and MMP-3 but not Chol/DOTAP liposomes or DC-Chol/DOPE liposomes. Taken together, we suggested that PC contained liposomes could provide the best liposomal formulation for the treatment of bone diseases.
Collapse
Affiliation(s)
- Shun-Fu Chang
- Department of Medical Research and Development, Chang Gung Memorial Hospital Chiayi Branch, Chiayi 61363, Taiwan.
| | - Chih-Chang Yeh
- Department of Orthopaedics, Chiayi Branch, Taichung Veterans General Hospital, No. 600, Sec. 2, Shixian Road, West District, Chiayi City 60090, Taiwan.
| | - Pin-Jyun Chen
- Department of Biochemical Science and Technology, National Chia Yi University, No. 300, Syuefu Rd, Chiayi City 60004, Taiwan.
| | - Hsin-I Chang
- Department of Biochemical Science and Technology, National Chia Yi University, No. 300, Syuefu Rd, Chiayi City 60004, Taiwan.
| |
Collapse
|
27
|
Chen G, Kawazoe N. Biomimetic Extracellular Matrices and Scaffolds Prepared from Cultured Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1078:465-474. [DOI: 10.1007/978-981-13-0950-2_24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
28
|
Abstract
Bone marrow fat cells comprise the largest population of cells in the bone marrow cavity, a characteristic that has attracted the attention of scholars from different disciplines. The perception that bone marrow adipocytes are "inert space fillers" has been broken, and currently, bone marrow fat is unanimously considered to be the third largest fat depot, after subcutaneous fat and visceral fat. Bone marrow fat (BMF) acts as a metabolically active organ and plays an active role in energy storage, endocrine function, bone metabolism, and the bone metastasis of tumors. Bone marrow adipocytes (BMAs), as a component of the bone marrow microenvironment, influence hematopoiesis through direct contact with cells and the secretion of adipocyte-derived factors. They also influence the progression of hematologic diseases such as leukemia, multiple myeloma, and aplastic anemia, and may be a novel target when exploring treatments for related diseases in the future. Based on currently available data, this review describes the role of BMF in hematopoiesis as well as in the development of hematologic diseases.
Collapse
|
29
|
Hoshiba T, Kawazoe N, Chen G. Preparation of Cell-Derived Decellularized Matrices Mimicking Native ECM During the Osteogenesis and Adipogenesis of Mesenchymal Stem Cells. Methods Mol Biol 2018; 1577:71-86. [PMID: 28795365 DOI: 10.1007/7651_2017_62] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The extracellular matrix (ECM) is an important extracellular microenvironmental factor that regulates stem cell differentiation. The ECM is remodeled according to stem cell differentiation progression to precisely regulate the differentiation. Thus, it is expected that the matrices mimicking native ECM surrounding differentiating cells at each differentiation stage provide a favorable microenvironment to promote stem cell differentiation. However, it is difficult to prepare matrices mimicking native ECM using chemical methods because the ECM has a complicated composition. The decellularization technique is useful to prepare such matrices. In this chapter, we described the protocol to prepare matrices mimicking native ECM surrounding cells that are differentiating from mesenchymal stem cells to either osteoblasts or adipocytes via stem cell differentiation culture and a detergent- and nuclease-based decellularization technique.
Collapse
Affiliation(s)
- Takashi Hoshiba
- Frontier Center for Organic Materials, Yamagata University, 4-3-16 Jonan, Yonezawa, Yamagata, 992-8510, Japan.
- Innovative Flex Course for Frontier Organic Materials Systems, Yamagata University, 4-3-16 Jonan, Yonezawa, Yamagata, 992-8510, Japan.
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.
| | - Naoki Kawazoe
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Guoping Chen
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| |
Collapse
|
30
|
Ma J, Guo W, Gao M, Huang B, Qi Q, Ling Z, Chen Y, Hu H, Zhou H, Yu F, Chen K, Richards G, Lin J, Zhou Z, Xiao D, Zou X. Biomimetic matrix fabricated by LMP-1 gene-transduced MC3T3-E1 cells for bone regeneration. Biofabrication 2017; 9:045010. [PMID: 28930090 DOI: 10.1088/1758-5090/aa8dd1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bone healing is regulated by multiple microenvironmental signals provided by the extracellular matrix (ECM). This study aimed to mimic the native osteoinductive microenvironment by developing an ECM using gene-transduced cells. The LIM mineralization protein-1 (LMP-1) gene was transferred to murine pre-osteoblast cells (MC3T3-E1) using lentiviral vectors. Western blotting assay indicated that the MC3T3-E1 cells expressed an increased level of bone morphologic protein-2, -4 and -7 (BMP-2, -4 and -7) after LMP-1 gene transduction. The transduced cells were then seeded into calcined bovine bone scaffolds and cultured for 7, 14, and 21 days to construct ECMs on the scaffolds. The ECM-scaffold composites were then decellularized using the freeze-drying method. Scaffolds without ECM deposition were used as controls. The composites and controls were implanted into critical-sized bone defects created in the distal femurs of New Zealand rabbits. Twelve weeks after the surgery, both microcomputed tomography and histologic results indicated that the 7-day-cell-modified ECM-scaffold composites induced bone regeneration with significantly larger volume, trabecular thickness and connectivity than the controls. However, the 14- and 21-day-cell-modified ECM-scaffold composites triggered sustained inflammation response even at 12 weeks after the surgery and showed less bone ingrowth and integration than their 7-day-cell-modified counterparts. In conclusion, these results highlight the viable gene transfer techniques for manipulating cells in a constructed microenvironment of ECM for bone regeneration. However, the unresolved inflammation relating to the duration of ECM modification needs to be considered.
Collapse
Affiliation(s)
- Junxuan Ma
- Department of Orthopedic, Peking University Shenzhen Hospital, Shenzhen, People's Republic of China. Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliate Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Basoli V, Santaniello S, Cruciani S, Ginesu GC, Cossu ML, Delitala AP, Serra PA, Ventura C, Maioli M. Melatonin and Vitamin D Interfere with the Adipogenic Fate of Adipose-Derived Stem Cells. Int J Mol Sci 2017; 18:ijms18050981. [PMID: 28475114 PMCID: PMC5454894 DOI: 10.3390/ijms18050981] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 04/28/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) represent one of the cellular populations resident in adipose tissue. They can be recruited under certain stimuli and committed to become preadipocytes, and then mature adipocytes. Controlling stem cell differentiation towards the adipogenic phenotype could have a great impact on future drug development aimed at counteracting fat depots. Stem cell commitment can be influenced by different molecules, such as melatonin, which we have previously shown to be an osteogenic inducer. Here, we aimed at evaluating the effects elicited by melatonin, even in the presence of vitamin D, on ADSC adipogenesis assessed in a specific medium. The transcription of specific adipogenesis orchestrating genes, such as aP2, peroxisome proliferator-activated receptor γ (PPAR-γ), and that of adipocyte-specific genes, including lipoprotein lipase (LPL) and acyl-CoA thioesterase 2 (ACOT2), was significantly inhibited in cells that had been treated in the presence of melatonin and vitamin D, alone or in combination. Protein content and lipid accumulation confirmed a reduction in adipogenesis in ADSCs that had been grown in adipogenic conditions, but in the presence of melatonin and/or vitamin D. Our findings indicate the role of melatonin and vitamin D in deciding stem cell fate, and disclose novel therapeutic approaches against fat depots.
Collapse
Affiliation(s)
- Valentina Basoli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Via Massarenti, 40138 Bologna, Italy.
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, A-1190 Vienna, Austria.
| | - Sara Santaniello
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Via Massarenti, 40138 Bologna, Italy.
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, A-1190 Vienna, Austria.
| | - Giorgio Carlo Ginesu
- Clinical and Experimental Medicine Department, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy.
| | - Maria Laura Cossu
- Clinical and Experimental Medicine Department, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy.
| | | | - Pier Andrea Serra
- Clinical and Experimental Medicine Department, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy.
- Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Via Massarenti, 40138 Bologna, Italy.
- Stem Wave Institute for Tissue Healing (SWITH), Gruppo VillaMaria and Ettore Sansavini Health Science Foundation, Via Provinciale per Cotignola 9, 48022 Lugo, Ravenna, Italy.
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Via Massarenti, 40138 Bologna, Italy.
- Stem Wave Institute for Tissue Healing (SWITH), Gruppo VillaMaria and Ettore Sansavini Health Science Foundation, Via Provinciale per Cotignola 9, 48022 Lugo, Ravenna, Italy.
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, 09042 Cagliari, Italy.
| |
Collapse
|
32
|
Teng Y, Li X, Chen Y, Cai H, Cao W, Chen X, Sun Y, Liang J, Fan Y, Zhang X. Extracellular matrix powder from cultured cartilage-like tissue as cell carrier for cartilage repair. J Mater Chem B 2017; 5:3283-3292. [PMID: 32264394 DOI: 10.1039/c7tb00640c] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Cartilage extracellular matrix (ECM) is a promising material for cartilage repair because of its bioactivity. However, the animal source of ECM unavoidably increases the risk of pathogen infection and the variability of product quality. In this study, we utilized a novel 3D culture method to prepare a new type of artificial decellularized matrix powder (DEMP) for the development of injectable, bioactive, biodegradable cell carriers for cartilage tissue engineering. This culture method combined hanging drop culture with suspension culture method, and was very efficient to produce cartilage-like tissue (CLT). By this method, an initial 2.3 × 106 chondrocyte generated as much as 58.22 mg wet weight CLT at two weeks, which proved to contain abundant glycoaminoglycans (GAGs), type II collagen, and BMP-2 and TGF-β1 growth factors by staining techniques and biochemical analysis. Subsequently, the two-week-old CLT was decellularized to prepare the artificial DEMP. In an in vitro study, it was found that MSCs cultured on DEMP differentiated to chondrocytes very well and secreted rich GAGs and type II collagen at three weeks even without exogenous TGF-β1. The in vivo study demonstrated that the DEMP not only facilitated regeneration of hyaline cartilage, which was implied by the intense staining of GAGs and type II collagen in rabbit subchondral defects at 1 month, but also benefited the regeneration of subchondral bone (bone ingrowth at 1 month: 48.22%) as shown in micro-CT data. Collectively, these results suggest that the artificial DEMP prepared by this culture method holds great potential as a novel ECM material for cartilage repair.
Collapse
Affiliation(s)
- Yingying Teng
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Role of RHEB in Regulating Differentiation Fate of Mesenchymal Stem Cells for Cartilage and Bone Regeneration. Int J Mol Sci 2017; 18:ijms18040880. [PMID: 28441755 PMCID: PMC5412461 DOI: 10.3390/ijms18040880] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/18/2022] Open
Abstract
Advances in mesenchymal stem cells (MSCs) and cell replacement therapies are promising approaches to treat cartilage and bone defects since substantial differentiation capacities of MSCs match the demands of tissue regeneration. Our understanding of the dynamic process requiring indispensable differentiation of MSCs remains limited. Herein, we describe the role of RHEB (Ras homolog enriched in brain) regulating gene signature for differentiation of human adipose derived mesenchymal stem cells (ASCs) into chondrogenic, osteogenic, and adipogenic lineages. RHEB-overexpression increases the proliferation of the ASCs. RHEB enhances the chondrogenic differentiation of ASCs in 3D culture via upregulation of SOX9 with concomitant increase in glycosaminoglycans (GAGs), and type II collagen (COL2). RHEB increases the osteogenesis via upregulation of runt related transcription factor 2 (RUNX2) with an increase in the calcium and phosphate contents. RHEB also increases the expression of osteogenic markers, osteonectin and osteopontin. RHEB knockdown ASCs were incapable of expressing sufficient SRY (Sex determining region Y)-box 9 (SOX9) and RUNX2, and therefore had decreased chondrogenic and osteogenic differentiation. RHEB-overexpression impaired ASCs differentiation into adipogenic lineage, through downregulation of CCAAT/enhancer binding protein beta (C/EBPβ). Conversely, RHEB knockdown abolished the negative regulation of adipogenesis. We demonstrate that RHEB is a novel regulator, with a critical role in ASCs lineage determination, and RHEB-modulated ASCs may be useful as a cell therapy for cartilage and bone defect treatments.
Collapse
|
34
|
Retinoic acid exacerbates chlorpyrifos action in ensuing adipogenic differentiation of C3H10T½ cells in a GSK3β dependent pathway. PLoS One 2017; 12:e0173031. [PMID: 28291828 PMCID: PMC5349446 DOI: 10.1371/journal.pone.0173031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 02/14/2017] [Indexed: 01/15/2023] Open
Abstract
The cell differentiation can be exploited as a paradigm to evaluate the effects of noxious chemicals, on human health, either alone or in combinations. In this regard, the effect of a known cell differentiation agent, retinoic acid (RA) was analyzed in the presence of a noxious chemical chlorpyrifos (CPF), an organophosphate (OP), the receptors of which have recently been localized to mesenchymal stem cells (MSCs). The observed imbalance of adipogenic to skeletal differentiation by CPF together with conundrum about adipogenic potential of RA prompted us to delineate their combinatorial effects on C3H10T½MSC-like undifferentiated cells. Based on MTT assay, the cellular viability was retained by CPF at concentrations ranging from 0.01–50μM, beyond which it caused cytotoxicity. These non-toxic concentrations also mildly interfered with adipogenesis of C3H10T½ cells following exposure to adipogenic cocktail. However, upon exposure to RA alone, these MSCs adopted elongated morphology and accumulated lipid vesicles, by day 20, as discerned by phase-contrast and transmission electron microscopy (TEM), in concert with enhanced Oil Red O stained cells. This effect got strongly augmented upon exposure to combination of CPF and RA in a dose-dependent manner. Simultaneous up-regulation in perilipin-1 (PLIN1) and adipsin (ADN) genes, additionally reiterated the adipogenic differentiation. Mechanistically, GSK3β pathway was found to be a major player, whereby inhibiting it with lithium chloride (LiCl) resulted in complete blockage of lipid accumulation, accompanied by complete down regulation of PLIN1 and ADN gene expression. In conclusion, these observations for the first time, lend evidence that exposure of CPF accompanied by RA directs commitment of C3H10T½ cells to adipogenic differentiation through a process involving a crosstalk at GSK3β signaling.
Collapse
|
35
|
Moeinzadeh S, Shariati SRP, Kader S, Melero-Martin JM, Jabbari E. Devitalized Stem Cell Microsheets for Sustainable Release of Osteogenic and Vasculogenic Growth Factors and Regulation of Anti-Inflammatory Immune Response. ACTA ACUST UNITED AC 2017; 1. [PMID: 30221188 DOI: 10.1002/adbi.201600011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The objective of this work was to investigate the effect of devitalized human mesenchymal stem cells (hMSCs) and endothelial colony-forming cells (ECFCs) seeded on mineralized nanofiber microsheets on protein release, osteogenesis, vasculogenesis, and macrophage polarization. Calcium phosphate nanocrystals were grown on the surface of aligned, functionalized nanofiber microsheets. The microsheets were seeded with hMSCs, ECFCs, or a mixture of hMSCs+ECFCs, cultured for cell attachment, differentiated to the osteogenic or vasculogenic lineage, and devitalized by lyophilization. The release kinetic of total protein, bone morphogenetic protein-2 (BMP2), and vascular endothelial growth factor (VEGF) from the devitalized microsheets was measured. Next, hMSCs and/or ECFCs were seeded on the devitalized cell microsheets and cultured in the absence of osteo-/vasculo-inductive factors to determine the effect of devitalized cell microsheets on hMSC/ECFC differentiation. Human macrophages were seeded on the microsheets to determine the effect of devitalized cells on macrophage polarization. Based on the results, devitalized undifferentiated hMSC and vasculogenic-differentiated ECFC microsheets had highest sustained release of BMP2 and VEGF, respectively. The devitalized hMSC microsheets did not affect M2 macrophage polarization while vascular-differentiated, devitalized ECFC microsheets did not affect M1 polarization. Both groups stimulated higher M2 macrophage polarization compared to M1.
Collapse
Affiliation(s)
- Seyedsina Moeinzadeh
- Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - Seyed Ramin Pajoum Shariati
- Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - Safaa Kader
- Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA.,Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Esmaiel Jabbari
- Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
36
|
Jafari A, Qanie D, Andersen TL, Zhang Y, Chen L, Postert B, Parsons S, Ditzel N, Khosla S, Johansen HT, Kjærsgaard-Andersen P, Delaisse JM, Abdallah BM, Hesselson D, Solberg R, Kassem M. Legumain Regulates Differentiation Fate of Human Bone Marrow Stromal Cells and Is Altered in Postmenopausal Osteoporosis. Stem Cell Reports 2017; 8:373-386. [PMID: 28162997 PMCID: PMC5312427 DOI: 10.1016/j.stemcr.2017.01.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/03/2017] [Accepted: 01/04/2017] [Indexed: 12/21/2022] Open
Abstract
Secreted factors are a key component of stem cell niche and their dysregulation compromises stem cell function. Legumain is a secreted cysteine protease involved in diverse biological processes. Here, we demonstrate that legumain regulates lineage commitment of human bone marrow stromal cells and that its expression level and cellular localization are altered in postmenopausal osteoporotic patients. As shown by genetic and pharmacological manipulation, legumain inhibited osteoblast (OB) differentiation and in vivo bone formation through degradation of the bone matrix protein fibronectin. In addition, genetic ablation or pharmacological inhibition of legumain activity led to precocious OB differentiation and increased vertebral mineralization in zebrafish. Finally, we show that localized increased expression of legumain in bone marrow adipocytes was inversely correlated with adjacent trabecular bone mass in a cohort of patients with postmenopausal osteoporosis. Our data suggest that altered proteolytic activity of legumain in the bone microenvironment contributes to decreased bone mass in postmenopausal osteoporosis. Legumain determines differentiation fate of BMSCs in vitro and in vivo Legumain regulates BMSC proliferation independent of its enzymatic activity Inhibition of legumain leads to precocious bone formation in zebrafish Legumain is overexpressed in bone marrow adipocytes of osteoporotic patients
Collapse
Affiliation(s)
- Abbas Jafari
- Department of Cellular and Molecular Medicine, Danish Stem Cell Center (DanStem), University of Copenhagen, 2200 Copenhagen, Denmark; Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark
| | - Diyako Qanie
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark
| | - Thomas L Andersen
- Department of Clinical Cell Biology, Vejle/ Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, 7100, Vejle, Denmark
| | - Yuxi Zhang
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Li Chen
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark
| | - Benno Postert
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Stuart Parsons
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Nicholas Ditzel
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark
| | - Sundeep Khosla
- Endocrine Research Unit, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | - Jean-Marie Delaisse
- Department of Clinical Cell Biology, Vejle/ Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, 7100, Vejle, Denmark
| | - Basem M Abdallah
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark; Department of Biological Sciences, College of Science, King Faisal University, Hofuf 6996, Saudi Arabia
| | - Daniel Hesselson
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St Vincent's Clinical School, UNSW Australia, Sydney, NSW 2010, Australia
| | - Rigmor Solberg
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, 0363 Oslo, Norway
| | - Moustapha Kassem
- Department of Cellular and Molecular Medicine, Danish Stem Cell Center (DanStem), University of Copenhagen, 2200 Copenhagen, Denmark; Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, J.B. Winsloewsvej 25, 1st Floor, 5000 Odense C, Denmark; Stem Cell Unit, Department of Anatomy, Faculty of Medicine, King Saud University, Riyadh 12372, Saudi Arabia.
| |
Collapse
|
37
|
Hoshiba T. Cultured cell-derived decellularized matrices: a review towards the next decade. J Mater Chem B 2017; 5:4322-4331. [DOI: 10.1039/c7tb00074j] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Summary of recent progress in cell-derived decellularized matrices preparation and application, with perspectives towards the next decade.
Collapse
Affiliation(s)
- T. Hoshiba
- Frontier Center for Organic Materials
- Yamagata University
- Yonezawa
- Japan
- Innovative Flex Course for Frontier Organic Materials Systems
| |
Collapse
|
38
|
Guneta V, Tan NS, Chan SKJ, Tanavde V, Lim TC, Wong TCM, Choong C. Comparative study of adipose-derived stem cells and bone marrow-derived stem cells in similar microenvironmental conditions. Exp Cell Res 2016; 348:155-164. [PMID: 27658569 DOI: 10.1016/j.yexcr.2016.09.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/20/2016] [Accepted: 09/18/2016] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs), which were first isolated from the bone marrow, are now being extracted from various other tissues in the body, including the adipose tissue. The current study presents systematic evidence of how the adipose tissue-derived stem cells (ASCs) and bone marrow-derived mesenchymal stem cells (Bm-MSCs) behave when cultured in specific pro-adipogenic microenvironments. The cells were first characterized and identified as MSCs in terms of their morphology, phenotypic expression, self-renewal capabilities and multi-lineage potential. Subsequently, the proliferation and gene expression profiles of the cell populations cultured on two-dimensional (2D) adipose tissue extracellular matrix (ECM)-coated tissue culture plastic (TCP) and in three-dimensional (3D) AlgiMatrix® microenvironments were analyzed. Overall, it was found that adipogenesis was triggered in both cell populations due to the presence of adipose tissue ECM. However, in 3D microenvironments, ASCs and Bm-MSCs were predisposed to the adipogenic and osteogenic lineages respectively. Overall, findings from this study will contribute to ongoing efforts in adipose tissue engineering as well as provide new insights into the role of the ECM and cues provided by the immediate microenvironment for stem cell differentiation.
Collapse
Affiliation(s)
- Vipra Guneta
- Division of Materials Technology, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Nguan Soon Tan
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; KK Research Centre, KK Women's and Children Hospital, 100 Bukit Timah Road, Singapore 229899, Singapore; Institute of Molecular and Cell Biology, Agency for Science Technology & Research (A⁎STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Soon Kiat Jeremy Chan
- School of Biological Science, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Vivek Tanavde
- Bioinformatics Institute, Agency for Science Technology & Research (A⁎STAR), 30 Biopolis Street, Matrix, Singapore 138671, Singapore
| | - Thiam Chye Lim
- Division of Plastic, Reconstructive and Aesthetic Surgery, Department of Surgery, National University Hospital (NUH) and National University of Singapore (NUS), Kent Ridge Wing, Singapore 119074, Singapore
| | - Thien Chong Marcus Wong
- Plastic, Reconstructive and Aesthetic Surgery Section, Tan Tock Seng Hospital (TTSH), 11, Jalan Tan Tock Seng, Singapore 308433, Singapore
| | - Cleo Choong
- Division of Materials Technology, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; KK Research Centre, KK Women's and Children Hospital, 100 Bukit Timah Road, Singapore 229899, Singapore.
| |
Collapse
|
39
|
Qiu X, Gui Y, Xu Y, Li D, Wang L. DHEA promotes osteoblast differentiation by regulating the expression of osteoblast-related genes and Foxp3(+) regulatory T cells. Biosci Trends 2016; 9:307-14. [PMID: 26559023 DOI: 10.5582/bst.2015.01073] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Several studies have reported that dehydroepiandrosterone (DHEA) promotes osteoblast proliferation and inhibits osteoblast apoptosis and that DHEA inhibits osteoclast maturation. However, whether DHEA regulates osteoblast differentiation remains unclear. The present study first examined the effect of DHEA on bone morphology in vivo. DHEA was found to increase bone volume (BV), bone mineral density (BMD), and the number of trabeculae in bone (Th.N) and it was found to decrease trabecular spacing in bone (Th.sp) in ovariectomized (OVX) mice. Next, the effect of DHEA on osteoblast differentiation was examined in vitro and osteoblastogenesis-related marker genes, such as Runx2, Osterix, Collagen1, and Osteocalcin, were also detected. DHEA increased osteoblast production in mesenchymal stem cells (MSCs) cultured in osteoblastogenic medium, and DHEA increased the expression of Runx2 and osterix, thereby increasing the expression of osteocalcin and collagen1. Immune cells and bone interact, so changes in immune cells were detected in vivo. DHEA increased the number of Foxp3(+) regulatory T cells (Tregs) in the spleen but it did not affect CTLA-4 or IL-10. When MSCs were treated with DHEA in the presence of Tregs, alkaline phosphatase (ALP) activity increased. Osteoblasts and adipocytes are both generated by MSCs. If osteoblast differentiation increases, adipocyte differentiation will decrease, and the reverse also holds true. DHEA was found to increase the number of adipocytes in osteoblastogenic medium but it had no effect on the number of adipocytes and expression of PPARγ mRNA in adipogenic medium. This finding suggests that osteoblasts may be involved in adipocyte production. In conclusion, the current results suggest that DHEA can improve postmenopausal osteoporosis (PMO) by up-regulating osteoblast differentiation via the up-regulation of the expression of osteoblastogenesis-related genes and via an increase in Foxp3(+) Tregs.
Collapse
Affiliation(s)
- Xuemin Qiu
- Obstetrics and Gynecology Hospital, Fudan University
| | | | | | | | | |
Collapse
|
40
|
Hoshiba T, Tanaka M. Decellularized matrices as in vitro models of extracellular matrix in tumor tissues at different malignant levels: Mechanism of 5-fluorouracil resistance in colorectal tumor cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2749-2757. [PMID: 27558478 DOI: 10.1016/j.bbamcr.2016.08.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 12/31/2022]
Abstract
Chemoresistance is a major barrier for tumor chemotherapy. It is well-known that chemoresistance increases with tumor progression. Chemoresistance is altered by both genetic mutations and the alteration of extracellular microenvironment. Particularly, the extracellular matrix (ECM) is remodeled during tumor progression. Therefore, ECM remodeling is expected to cause the acquisition of chemoresistance in highly malignant tumor tissue. Here, we prepared cultured cell-derived decellularized matrices that mimic native ECM in tumor tissues at different stages of malignancy, and 5-fluorouracil (5-FU) resistance was compared among these matrices. 5-FU resistance of colorectal tumor cells increased on the matrices derived from highly malignant tumor HT-29 cells, although the resistance did not increase on the matrices derived from low malignant tumor SW480 cells and normal CCD-841-CoN cells. The resistance on HT-29 cell-derived matrices increased through the activation of Akt and the upregulation of ABCB1 and ABCC1 without cell growth promotion, suggesting that ECM remodeling plays important roles in the acquisition of chemoresistance during tumor progression. It is expected that our decellularized matrices, or "staged tumorigenesis-mimicking matrices", will become preferred cell culture substrates for in vitro analysis of comprehensive ECM roles in chemoresistance and the screening and pharmacokinetic analysis of anti-cancer drugs.
Collapse
Affiliation(s)
- Takashi Hoshiba
- Frontier Center for Organic Materials, Yamagata University, 4-3-16 Jonan, Yonezawa, Yamagata 992-8510, Japan; International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.
| | - Masaru Tanaka
- Frontier Center for Organic Materials, Yamagata University, 4-3-16 Jonan, Yonezawa, Yamagata 992-8510, Japan; Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, Fukuoka 819-0395, Japan
| |
Collapse
|
41
|
Deng P, Chen QM, Hong C, Wang CY. Histone methyltransferases and demethylases: regulators in balancing osteogenic and adipogenic differentiation of mesenchymal stem cells. Int J Oral Sci 2015; 7:197-204. [PMID: 26674421 PMCID: PMC5153596 DOI: 10.1038/ijos.2015.41] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2015] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are characterized by their self-renewing capacity and differentiation potential into multiple tissues. Thus, management of the differentiation capacities of MSCs is important for MSC-based regenerative medicine, such as craniofacial bone regeneration, and in new treatments for metabolic bone diseases, such as osteoporosis. In recent years, histone modification has been a growing topic in the field of MSC lineage specification, in which the Su(var)3–9, enhancer-of-zeste, trithorax (SET) domain-containing family and the Jumonji C (JmjC) domain-containing family represent the major histone lysine methyltransferases (KMTs) and histone lysine demethylases (KDMs), respectively. In this review, we summarize the current understanding of the epigenetic mechanisms by which SET domain-containing KMTs and JmjC domain-containing KDMs balance the osteogenic and adipogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Peng Deng
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA.,State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Qian-Ming Chen
- State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Christine Hong
- Section of Orthodontics, School of Dentistry, University of California at Los Angeles, Los Angeles, USA
| | - Cun-Yu Wang
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California at Los Angeles, Los Angeles, USA
| |
Collapse
|
42
|
Poulos SP, Dodson MV, Culver MF, Hausman GJ. The increasingly complex regulation of adipocyte differentiation. Exp Biol Med (Maywood) 2015; 241:449-56. [PMID: 26645953 DOI: 10.1177/1535370215619041] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/29/2015] [Indexed: 01/31/2023] Open
Abstract
Adipose (AD) tissue development and function relies on the ability of adipocytes to proliferate and differentiate into lipid-containing cells that also have endocrine function. Research suggests that certain conditions can induce AD tissue stem cells to differentiate into various cell types and that the microenvironment of the cell, including the extracellular matrix (ECM), is essential in maintaining cell and tissue function. This review provides an overview of factors involved in the proliferation and differentiation of adipocytes. A brief review of the numerous factors that influence PPARγ, the transcription factor thought to be the master regulator of adipocyte differentiation, provides context of established pathways that regulate adipogenesis. Thought provoking findings from research with hypoxia that is supported by earlier research that vascular development is related to adipogenesis are reviewed. Finally, our understanding of the critical role of the ECM and environment in adipogenesis is discussed and compared with studies that suggest that adipocytes may dedifferentiate and can convert into other cell types.
Collapse
Affiliation(s)
| | - Michael V Dodson
- Department of Animal Science, Washington State University, Pullman, WA 99164, USA
| | | | - Gary J Hausman
- Animal and Dairy Science Department, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
43
|
Hoshiba T, Chen G, Endo C, Maruyama H, Wakui M, Nemoto E, Kawazoe N, Tanaka M. Decellularized Extracellular Matrix as an In Vitro Model to Study the Comprehensive Roles of the ECM in Stem Cell Differentiation. Stem Cells Int 2015; 2016:6397820. [PMID: 26770210 PMCID: PMC4684892 DOI: 10.1155/2016/6397820] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/30/2015] [Indexed: 12/16/2022] Open
Abstract
Stem cells are a promising cell source for regenerative medicine. Stem cell differentiation must be regulated for applications in regenerative medicine. Stem cells are surrounded by extracellular matrix (ECM) in vivo. The ECM is composed of many types of proteins and glycosaminoglycans that assemble into a complex structure. The assembly of ECM molecules influences stem cell differentiation through orchestrated intracellular signaling activated by many ECM molecules. Therefore, it is important to understand the comprehensive role of the ECM in stem cell differentiation as well as the functions of the individual ECM molecules. Decellularized ECM is a useful in vitro model for studying the comprehensive roles of ECM because it retains a native-like structure and composition. Decellularized ECM can be obtained from in vivo tissue ECM or ECM fabricated by cells cultured in vitro. It is important to select the correct decellularized ECM because each type has different properties. In this review, tissue-derived and cell-derived decellularized ECMs are compared as in vitro ECM models to examine the comprehensive roles of the ECM in stem cell differentiation. We also summarize recent studies using decellularized ECM to determine the comprehensive roles of the ECM in stem cell differentiation.
Collapse
Affiliation(s)
- Takashi Hoshiba
- Frontier Center for Organic Materials, Yamagata University, 4-3-16 Jonan, Yonezawa, Yamagata 992-8510, Japan
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Guoping Chen
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Chiho Endo
- Graduate School of Science and Engineering, Yamagata University, 4-3-16 Jonan, Yonezawa, Yamagata 992-8510, Japan
| | - Hiroka Maruyama
- Graduate School of Science and Engineering, Yamagata University, 4-3-16 Jonan, Yonezawa, Yamagata 992-8510, Japan
| | - Miyuki Wakui
- Graduate School of Science and Engineering, Yamagata University, 4-3-16 Jonan, Yonezawa, Yamagata 992-8510, Japan
| | - Eri Nemoto
- Graduate School of Science and Engineering, Yamagata University, 4-3-16 Jonan, Yonezawa, Yamagata 992-8510, Japan
| | - Naoki Kawazoe
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Masaru Tanaka
- Frontier Center for Organic Materials, Yamagata University, 4-3-16 Jonan, Yonezawa, Yamagata 992-8510, Japan
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, Fukuoka 819-0395, Japan
| |
Collapse
|
44
|
Ko DY, Patel M, Jung BK, Park JH, Jeong B. Phosphorylcholine-Based Zwitterionic Biocompatible Thermogel. Biomacromolecules 2015; 16:3853-62. [DOI: 10.1021/acs.biomac.5b01169] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Du Young Ko
- Department of Chemistry and
Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 120-750, Korea
| | - Madhumita Patel
- Department of Chemistry and
Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 120-750, Korea
| | - Bo Kyoeng Jung
- Department of Chemistry and
Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 120-750, Korea
| | - Jin Hye Park
- Department of Chemistry and
Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 120-750, Korea
| | - Byeongmoon Jeong
- Department of Chemistry and
Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 120-750, Korea
| |
Collapse
|
45
|
Liu HY, Huang CF, Lin TC, Tsai CY, Tina Chen SY, Liu A, Chen WH, Wei HJ, Wang MF, Williams DF, Deng WP. Delayed animal aging through the recovery of stem cell senescence by platelet rich plasma. Biomaterials 2014; 35:9767-9776. [DOI: 10.1016/j.biomaterials.2014.08.034] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/21/2014] [Indexed: 11/28/2022]
|
46
|
Zhao J, Wang C, Song Y, Fang B. Arsenic trioxide and microRNA-204 display contrary effects on regulating adipogenic and osteogenic differentiation of mesenchymal stem cells in aplastic anemia. Acta Biochim Biophys Sin (Shanghai) 2014; 46:885-93. [PMID: 25187411 DOI: 10.1093/abbs/gmu082] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Our previous studies have demonstrated that arsenic trioxide (ATO) had the clinical efficacy in treating patients with aplastic anemia (AA). However, the mechanisms remain to be elucidated. The important components of the bone marrow hematopoietic microenvironment, bone marrow mesenchymal stem cells (BMSCs), are often altered in AA patients. In this study, it was found that AA BMSCs were prone to be induced into adipocytes rather than osteoblasts. ATO treatment can at least partially restore the differentiation imbalance of AA BMSCs. We further identified miR-204 as a key regulator in AA BMSC differentiation. Luciferase reporter assay showed that miR-204 could directly bind to the 3'-untranslated region of Runx2 mRNA, a key transcription factor regulating osteogenesis. Moreover, adipogenic differentiation was promoted and osteogenic differentiation was inhibited in miR-204 over-expressed cells, whereas osteogenesis was enhanced and adipocyte formation was inhibited in cells that lost miR-204 function, which suggested its endogenous function. Together we showed that ATO could inhibit adipogenic differentiation, but promote osteogenic differentiation in AA BMSCs, providing a possible explanation for ATO clinical efficacy in AA patients. MiR-204 plays a key role in regulating BMSCs differentiation, and down-regulating miR-204 expression might be a novel strategy to treat AA.
Collapse
Affiliation(s)
- Junmei Zhao
- Henan Key Lab of Experimental Haematology, Henan Institute of Haematology, Henan Tumor Hospital affiliated to Zhengzhou University, Zhengzhou 450008, China
| | - Chao Wang
- Henan Key Lab of Experimental Haematology, Henan Institute of Haematology, Henan Tumor Hospital affiliated to Zhengzhou University, Zhengzhou 450008, China
| | - Yongping Song
- Henan Key Lab of Experimental Haematology, Henan Institute of Haematology, Henan Tumor Hospital affiliated to Zhengzhou University, Zhengzhou 450008, China
| | - Baijun Fang
- Henan Key Lab of Experimental Haematology, Henan Institute of Haematology, Henan Tumor Hospital affiliated to Zhengzhou University, Zhengzhou 450008, China
| |
Collapse
|
47
|
Veronesi F, Pagani S, Della Bella E, Giavaresi G, Fini M. Estrogen deficiency does not decrease the in vitro osteogenic potential of rat adipose-derived mesenchymal stem cells. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9647. [PMID: 24687841 PMCID: PMC4082606 DOI: 10.1007/s11357-014-9647-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 03/18/2014] [Indexed: 05/13/2023]
Abstract
Osteoporosis due to estrogen deficiency is an increasing bone health issue worldwide: new strategies are being studied for regenerative medicine of bone pathologies in these patients. The most commonly used cells for tissue engineering therapy are the bone marrow mesenchymal stem cells (BMSCs), but they might be negatively affected by aging and estrogen deficiency. Besides the general advantages of adipose-derived mesenchymal stem cells (ADSCs) over BMSCs, ADSCs also seem to be less affected by aging than BMSCs, but in the literature, little is known about ADSCs in estrogen deficiency. The present study investigated the in vitro behavior of ADSCs, isolated from healthy (SHAM) and estrogen-deficient (OVX) rats. Phenotype, clonogenicity, viability, and osteogenic differentiation, at both cellular and molecular levels, were evaluated with or without osteogenic stimuli. Pro-inflammatory cytokines, growth factors, and adipogenic differentiation markers were also analyzed. There were no significant differences between OVX and SHAM ADSCs in some analyzed parameters. In addition, clonogenicity, osteopontin (Spp1) gene expression, alkaline phosphatase (ALP) activity at 2 weeks of culture, total collagen (COLL), osteocalcin (Bglap) gene expression and production, and matrix mineralization were significantly higher in OVX than in SHAM ADSCs. Besides the increase in some osteogenic markers, peroxisome proliferator-activated receptor gamma (Pparg) gene was also more expressed in OVX in osteogenic medium, with a concomitant estrogen receptor 1 (Esr1) gene expression decrease. These results underlined that ADSCs were not affected by estrogen deficiency in an osteogenic microenvironment.
Collapse
Affiliation(s)
- Francesca Veronesi
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, Via Di Barbiano 1/10, 40136, Bologna, Italy,
| | | | | | | | | |
Collapse
|
48
|
Li S, Wang M, Chen X, Li SF, Li-Ling J, Xie HQ. Inhibition of osteogenic differentiation of mesenchymal stem cells by copper supplementation. Cell Prolif 2014; 47:81-90. [PMID: 24450813 DOI: 10.1111/cpr.12083] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 09/21/2013] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Copper has been added to scaffolds when investigating bone repair, as an agent to promote vascularization; however, little is known concerning its effect on mesenchymal stem cells (MSCs), which are considered to be the origin of osteoblasts. In this study, we have aimed to elucidate effects of copper on osteogenic differentiation of MSCs. MATERIALS AND METHODS Rat bone marrow MSCs (rBMSCs) were used as a model. Their viability was assessed by MTT assay and Roche's CASY cell counter test and calcium deposition was evaluated by staining with alizarin red S. Fluorescent phalloidin F-actin stain was used to evaluate cytoskeletal changes, protein expressions were investigated by western blotting and mRNA levels were analysed using Q-PCR. A rat model for ectopic bone formation was used to assess effects of copper on MSCs in vivo. RESULTS Copper supplementation resulted in inhibition of osteogenesis of rBMSCs, along with reduction in expression of a number of osteogenic genes, alkaline phosphatase activity and formation of bone nodules. Cytoskeletal changes to cells during osteogenesis was inhibited by copper supplementation. In vivo study confirmed that copper could inhibit collagen formation whilst promoting angiogenesis. CONCLUSIONS Our study demonstrated that copper inhibited osteogenic differentiation of rBMSCs in vitro. The findings caution appropriate use of copper and have laid a foundation for further research.
Collapse
Affiliation(s)
- S Li
- Laboratory of Stem Cell and Tissue Engineering, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | | | | | | | | | | |
Collapse
|
49
|
Lorda-Diez CI, Montero JA, Choe S, Garcia-Porrero JA, Hurle JM. Ligand- and stage-dependent divergent functions of BMP signaling in the differentiation of embryonic skeletogenic progenitors in vitro. J Bone Miner Res 2014; 29:735-48. [PMID: 24038612 DOI: 10.1002/jbmr.2077] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/16/2013] [Accepted: 08/02/2013] [Indexed: 12/21/2022]
Abstract
Bone morphogenetic proteins (BMPs) are key molecules in the differentiation of skeletal tissues. We have investigated whether differentiation of limb embryonic mesodermal progenitors into different connective tissue lineages depends on specific stimulation of distinct BMP ligands or on the differential response of target cells to a common BMP stimulus. We show that Bmp2,4,5,7 and Gdf5 exhibit differential expression domains during the formation of tendons, cartilages, and joint tissues in digit development, but their respective effects on digit progenitors cell cultures cannot sustain the divergent differentiation of these cells into tendons, joints, and cartilage. However, the influence of BMPs differs based on the culture length. Early cultures respond to any of the BMPs by inducing chondrogenic factors and inhibiting fibrogenic and osteogenic markers. Later, a second phase of the culture occurs when BMPs attenuate their prochondrogenic influence and promote the fibrogenic marker Scleraxis. At advanced culture stages, BMPs inhibit prochondrogenic and profibrogenic markers and promote osteogenic markers. The switch from the prochondrogenic to the profibrogenic response appears critically dependent on the basal expression of Noggin. Thus, the differential regulation of Scleraxis at these stages was abrogated by treatments with a BMP-analogous compound (AB204) that escapes NOGGIN antagonism. Gene regulation experiments in absence of protein synthesis during the first period of culture indicate that BMPs activate at the same time master chondrogenic and fibrogenic genes together with cofactors responsible for driving the signaling cascade toward chondrogenesis or fibrogenesis. Gene-silencing experiments indicate that Id2 is one of the factors limiting the profibrogenic influence of BMPs. We propose that connective tissues are dynamic structures composed of cartilage, fibrous tissue, and bone that form in successive steps from the differentiation of common progenitors. This sequential differentiation is regulated by BMPs through a process that is dependent on the basal expression of BMP cofactors or signaling modulators.
Collapse
Affiliation(s)
- Carlos I Lorda-Diez
- Departamento de Anatomía y Biología Celular and IFIMAV, Universidad de Cantabria, Santander, Spain
| | | | | | | | | |
Collapse
|
50
|
Joddar B, Hoshiba T, Chen G, Ito Y. Stem cell culture using cell-derived substrates. Biomater Sci 2014; 2:1595-1603. [DOI: 10.1039/c4bm00126e] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
There have been great efforts to develop cell culture systems using chemically-fixed cells or decellularized matrices to regulate stem cell functions.
Collapse
Affiliation(s)
| | - Takashi Hoshiba
- Department of Biochemical Engineering
- Graduate School of Science and Engineering
- Yamagata University
- Yonezawa, Japan
- Tissue Regeneration Materials Unit
| | - Guoping Chen
- Tissue Regeneration Materials Unit
- International Center for Materials Nanoarchitectonics
- National Institute for Materials Science
- Tsukuba, Japan
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory
- RIKEN
- Wako, Japan
- Emergent Bioengineering Materials Research Team
- RIKEN Center for Emergent Matter Science
| |
Collapse
|