1
|
Sabatini C, Lin HJ, Ovik G, Hall R, Lee T. The proneural transcription factor Atoh1 promotes odontogenic differentiation in human dental pulp stem cells (DPSCs). BMC Mol Cell Biol 2025; 26:5. [PMID: 39833721 PMCID: PMC11744864 DOI: 10.1186/s12860-025-00530-2] [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: 10/18/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Bioengineering of human teeth for replacement is an appealing regenerative approach in the era of gene therapy. Developmentally regulated transcription factors hold promise in the quest because these transcriptional regulators constitute the gene regulatory networks driving cell fate determination. Atonal homolog 1 (Atoh1) is a transcription factor of the basic helix-loop-helix (bHLH) family essential for neurogenesis in the cerebellum, auditory hair cell differentiation, and intestinal stem cell specification. The functional versatility of Atoh1 prompted us to test the possibility that Atoh1 may intersect the dental pulp stem cell (DPSC) gene regulatory network governing odontogenic differentiation. METHODS We isolated DPSCs from human dental pulps and treated the cells with a replication-deficient adenoviral vector to achieve robust ectopic expression of Atoh1, following which the growth and odontogenic differentiation profiles of DPSCs were characterized. RESULTS DPSCs harboring the Atoh1 expression vector exhibited an approximately 3,000-fold increase in the expression of Atoh1 compared to the negative control, leading to increased DPSC proliferation in the growth medium (P < 0.05). In the odontogenic medium, Atoh1 caused an early induction of BMP2 (P < 0.001) followed by a late induction of BMP7 (P < 0.01) and increased Wnt signaling (P < 0.01). The increased BMP/Wnt signaling led to up to 8-fold increased expression of the master osteogenic transcription factor Osterix (P < 0.005) while exhibiting no significant effect on Runx2 or Dlx5, which are abundantly expressed in DPSCs. Atoh1 stimulated expression of type I collagen (P < 0.005) and small integrin-binding ligand, N-linked glycoproteins (SIBLINGs) such as bone sialoprotein (P < 0.001), dentin matrix protein 1 (P < 0.05), dentin sialophosphoprotein (P < 0.005), and osteopontin (P < 0.001), resulting in increased dentin matrix mineralization (P < 0.05). The odontogenic phenotype is associated with metabolic remodeling marked by enhanced glycolytic flux and attenuated mitochondrial metabolic enzyme activities. CONCLUSIONS Atoh1, despite being a proneural transcription factor in development, possesses a novel odontogenic function upon ectopic expression in DPSCs. This in vitro study demonstrates a novel odontogenic mechanism mediated by ectopic expression of the transcription factor Atoh1 in human DPSCs. The finding may offer an innovative strategy for gene-based regeneration of the pulp-dentin complex.
Collapse
Affiliation(s)
- Camila Sabatini
- Department of Restorative Dentistry, School of Dental Medicine, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA.
| | - Huey-Jiun Lin
- Department of Biochemistry, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Galib Ovik
- Department of Biochemistry, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Richard Hall
- Department of Oral Surgery, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Techung Lee
- Department of Biochemistry, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| |
Collapse
|
2
|
Chen C, Chen B, Lin Y, He Q, Yang J, Xiao J, Pan Z, Li S, Li M, Wang F, Zhang H, Wang X, Zeng J, Chi W, Meng K, Wang H, Chen P. Cardamonin attenuates iron overload-induced osteoblast oxidative stress through the HIF-1α/ROS pathway. Int Immunopharmacol 2024; 142:112893. [PMID: 39217878 DOI: 10.1016/j.intimp.2024.112893] [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: 05/28/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Osteoporosis(OP) is a bone disease under research. Iron overload is a significant risk factor. Iron balance is crucial for bone metabolism and biochemical processes. When there is an excess of iron in the body, it tends to produce reactive oxygen species (ROS) which can cause oxidative damage to cells. The flavonoid compound, Cardamonin (CAR), possesses potent anti-inflammatory and anti-iron overload properties that can be beneficial in mitigating the risk of OP. PURPOSE This study investigates the potential therapeutic interventions and underlying mechanisms of CAR for treating OP in individuals with iron overload. METHODS The model of iron-overloaded mice was established by intraperitoneally injecting iron dextran(ID) into the mice. OP severity was evaluated with micro-CT and Hematoxylin-Eosin (HE) staining in vivo. In vitro, the iron-overloaded osteoblast model was induced by ferric ammonium citrate. Cell counting kit 8 assay to evaluate cell viability, Annexin V-FITC/PI assay to detect cell apoptosis. A range of cellular markers were detected, including the variation in mitochondrial membrane potential (MMP), levels of malondialdehyde (MDA), ROS, and lipid hydroperoxide (LPO). RESULTS CAR can reverse bone loss in iron overload-induced OP mouse models in vivo. CAR attenuates the impairment of iron overload on the activity and apoptosis of MC3T3-E1 cells as well as the accumulation of ROS and LPO activation via HIF-1α/ROS pathways. CONCLUSION CAR downregulating HIF-1α pathways prevents inhibition of iron overload-induced osteoblasts dysfunctional by attenuating ROS accumulation, reducing oxidative stress, promotes bone formation, and alleviates OP.
Collapse
Affiliation(s)
- Chuyi Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, PR China; 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Bohao Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, PR China; 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Yuewei Lin
- 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qi He
- 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Junzheng Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, PR China; 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Jiacong Xiao
- 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Zhaofeng Pan
- 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Shaocong Li
- 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Miao Li
- 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Fanchen Wang
- 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Hua Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, PR China; 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Xintian Wang
- 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Jiaxu Zeng
- 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Weijin Chi
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, PR China; 1st School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Kai Meng
- Department of Orthopaedics Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, PR China.
| | - Haibin Wang
- Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China.
| | - Peng Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, PR China; Department of Orthopaedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou 510405, PR China; Xinjiang Production and Construction Corps 13th Division Red Star Hospital, Hami 839000, PR China; The Affiliated Redstar Hospital of Shihezi University School of Medicine, 832000, PR China.
| |
Collapse
|
3
|
Tian K, He X, Lin X, Chen X, Su Y, Lu Z, Chen Z, Zhang L, Li P, Ma L, Lan Z, Zhao X, Fen G, Hai Q, Xue D, Jin Q. Unveiling the Role of Sik1 in Osteoblast Differentiation: Implications for Osteoarthritis. Mol Cell Biol 2024; 44:411-428. [PMID: 39169784 PMCID: PMC11485870 DOI: 10.1080/10985549.2024.2385633] [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: 03/25/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease characterized by subchondral osteosclerosis, mainly due to osteoblast activity. This research investigates the function of Sik1, a member of the AMP-activated protein kinase family, in OA. Proteomic analysis was conducted on clinical samples from 30 OA patients, revealing a negative correlation between Sik1 expression and OA. In vitro experiments utilized BMSCs to examine the effect of Sik1 on osteogenic differentiation. BMSCs were cultured and induced toward osteogenesis with specific media. Sik1 overexpression was achieved through lentiviral transfection, followed by analysis of osteogenesis-associated proteins using Western blotting, RT-qPCR, and alkaline phosphate staining. In vivo experiments involved destabilizing the medial meniscus in mice to establish an OA model, assessing the therapeutic potential of Sik1. The CT scans and histological staining were used to analyze subchondral bone alterations and cartilage damage. The findings show that Sik1 downregulation correlates with advanced OA and heightened osteogenic differentiation in BMSCs. Sik1 overexpression inhibits osteogenesis-related markers in vitro and reduces cartilage damage and subchondral osteosclerosis in vivo. Mechanistically, Sik1 modulates osteogenesis and subchondral bone changes through Runx2 activity regulation. The research emphasizes Sik1 as a promising target for treating OA, suggesting its involvement in controlling bone formation and changes in the subchondral osteosclerosis.
Collapse
Affiliation(s)
- Kuanmin Tian
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xiaoxin He
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xue Lin
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xiaolei Chen
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yajing Su
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Zhidong Lu
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Zhirong Chen
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Liang Zhang
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Peng Li
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Long Ma
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Zhibin Lan
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xin Zhao
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Gangning Fen
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qinqin Hai
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Di Xue
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qunhua Jin
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| |
Collapse
|
4
|
Marom R, Song IW, Busse EC, Washington ME, Berrier AS, Rossi VC, Ortinau L, Jeong Y, Jiang MM, Dawson BC, Adeyeye M, Leynes C, Lietman CD, Stroup BM, Batkovskyte D, Jain M, Chen Y, Cela R, Castellon A, Tran AA, Lorenzo I, Meyers DN, Huang S, Turner A, Shenava V, Wallace M, Orwoll E, Park D, Ambrose CG, Nagamani SC, Heaney JD, Lee BH. The IFITM5 mutation in osteogenesis imperfecta type V is associated with an ERK/SOX9-dependent osteoprogenitor differentiation defect. J Clin Invest 2024; 134:e170369. [PMID: 38885336 PMCID: PMC11290974 DOI: 10.1172/jci170369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Osteogenesis imperfecta (OI) type V is the second most common form of OI, distinguished by hyperplastic callus formation and calcification of the interosseous membranes, in addition to the bone fragility. It is caused by a recurrent, dominant pathogenic variant (c.-14C>T) in interferon-induced transmembrane protein 5 (IFITM5). Here, we generated a conditional Rosa26-knockin mouse model to study the mechanistic consequences of the recurrent mutation. Expression of the mutant Ifitm5 in osteo-chondroprogenitor or chondrogenic cells resulted in low bone mass and growth retardation. Mutant limbs showed impaired endochondral ossification, cartilage overgrowth, and abnormal growth plate architecture. The cartilage phenotype correlates with the pathology reported in patients with OI type V. Surprisingly, expression of mutant Ifitm5 in mature osteoblasts caused no obvious skeletal abnormalities. In contrast, earlier expression in osteo-chondroprogenitors was associated with an increase in the skeletal progenitor cell population within the periosteum. Lineage tracing showed that chondrogenic cells expressing the mutant Ifitm5 had decreased differentiation into osteoblastic cells in diaphyseal bone. Moreover, mutant IFITM5 disrupted early skeletal homeostasis in part by activating ERK signaling and downstream SOX9 protein, and inhibition of these pathways partially rescued the phenotype in mutant animals. These data identify the contribution of a signaling defect altering osteo-chondroprogenitor differentiation as a driver in the pathogenesis of OI type V.
Collapse
Affiliation(s)
- Ronit Marom
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Houston, Texas, USA
| | - I-Wen Song
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Emily C. Busse
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, USA
| | - Megan E. Washington
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Ava S. Berrier
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Vittoria C. Rossi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Houston, Texas, USA
| | - Laura Ortinau
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Youngjae Jeong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Ming-Ming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Brian C. Dawson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Mary Adeyeye
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Medical Scientist Training Program, UT Health Houston MD Anderson Cancer Center, Houston, Texas, USA
| | - Carolina Leynes
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Caressa D. Lietman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Bridget M. Stroup
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Dominyka Batkovskyte
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Mahim Jain
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Yuqing Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Racel Cela
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Alexis Castellon
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Alyssa A. Tran
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Isabel Lorenzo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - D. Nicole Meyers
- Department of Orthopaedic Surgery, McGovern Medical School at UT Health, Houston, Texas, USA
| | - Shixia Huang
- Department of Molecular and Cellular Biology, and Huffington Department of Education, Innovation, and Technology, Advanced Technology Cores, and
| | - Alicia Turner
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Houston, Texas, USA
| | - Vinitha Shenava
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Maegen Wallace
- Orthopaedic Surgery, University of Nebraska Medical Center, Children’s Hospital and Medical Center, Omaha, Nebraska, USA
| | - Eric Orwoll
- Department of Medicine, Bone and Mineral Unit, Oregon Health and Science University, Portland, Oregon, USA
| | - Dongsu Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Catherine G. Ambrose
- Department of Orthopaedic Surgery, McGovern Medical School at UT Health, Houston, Texas, USA
| | - Sandesh C.S. Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Houston, Texas, USA
| | - Jason D. Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Brendan H. Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
5
|
Arambula-Maldonado R, Mequanint K. Osteogenic Differentiation Potential of iMSCs on GelMA-BG-MWCNT Nanocomposite Hydrogels. Biomimetics (Basel) 2024; 9:338. [PMID: 38921218 PMCID: PMC11201442 DOI: 10.3390/biomimetics9060338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
The ability of bone biomaterials to promote osteogenic differentiation is crucial for the repair and regeneration of osseous tissue. The development of a temporary bone substitute is of major importance in enhancing the growth and differentiation of human-derived stem cells into an osteogenic lineage. In this study, nanocomposite hydrogels composed of gelatin methacryloyl (GelMA), bioactive glass (BG), and multiwall carbon nanotubes (MWCNT) were developed to create a bone biomaterial that mimics the structural and electrically conductive nature of bone that can promote the differentiation of human-derived stem cells. GelMA-BG-MWCNT nanocomposite hydrogels supported mesenchymal stem cells derived from human induced pluripotent stem cells, hereinafter named iMSCs. Cell adhesion was improved upon coating nanocomposite hydrogels with fibronectin and was further enhanced when seeding pre-differentiated iMSCs. Osteogenic differentiation and mature mineralization were promoted in GelMA-BG-MWCNT nanocomposite hydrogels and were most evidently observed in the 70-30-2 hydrogels, which could be due to the stiff topography characteristic from the addition of MWCNT. Overall, the results of this study showed that GelMA-BG-MWCNT nanocomposite hydrogels coated with fibronectin possessed a favorable environment in which pre-differentiated iMSCs could better attach, proliferate, and further mature into an osteogenic lineage, which was crucial for the repair and regeneration of bone.
Collapse
Affiliation(s)
- Rebeca Arambula-Maldonado
- School of Biomedical Engineering, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B9, Canada;
| | - Kibret Mequanint
- School of Biomedical Engineering, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B9, Canada;
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, 1151 Richmond Street, London, ON N6A 5B9, Canada
| |
Collapse
|
6
|
Binlateh T, Leethanakul C, Thammanichanon P. Involvement of RAMP1/p38MAPK signaling pathway in osteoblast differentiation in response to mechanical stimulation: a preliminary study. J Orthop Surg Res 2024; 19:330. [PMID: 38825686 PMCID: PMC11145863 DOI: 10.1186/s13018-024-04805-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/21/2024] [Indexed: 06/04/2024] Open
Abstract
OBJECTIVE The present study aimed to investigate the underlying mechanism of mechanical stimulation in regulating osteogenic differentiation. MATERIALS AND METHODS Osteoblasts were exposed to compressive force (0-4 g/cm2) for 1-3 days or CGRP for 1 or 3 days. Expression of receptor activity modifying protein 1 (RAMP1), the transcription factor RUNX2, osteocalcin, p38 and p-p38 were analyzed by western blotting. Calcium mineralization was analyzed by alizarin red straining. RESULTS Using compressive force treatments, low magnitudes (1 and 2 g/cm2) of compressive force for 24 h promoted osteoblast differentiation and mineral deposition whereas higher magnitudes (3 and 4 g/cm2) did not produce osteogenic effect. Through western blot assay, we observed that the receptor activity-modifying protein 1 (RAMP1) expression was upregulated, and p38 mitogen-activated protein kinase (MAPK) was phosphorylated during low magnitudes compressive force-promoted osteoblast differentiation. Further investigation of a calcitonin gene-related peptide (CGRP) peptide incubation, a ligand for RAMP1, showed that CGRP at concentration of 25 and 50 ng/ml could increase expression levels of RUNX2 and osteocalcin, and percentage of mineralization, suggesting its osteogenic potential. In addition, with the same conditions, CGRP also significantly upregulated RAMP1 and phosphorylated p38 expression levels. Also, the combination of compressive forces (1 and 2 g/cm2) with 50 ng/ml CGRP trended to increase RAMP1 expression, p38 activity, and osteogenic marker RUNX2 levels, as well as percentage of mineralization compared to compressive force alone. This suggest that RAMP1 possibly acts as an upstream regulator of p38 signaling during osteogenic differentiation. CONCLUSION These findings suggest that CGRP-RAMP1/p38MAPK signaling implicates in osteoblast differentiation in response to optimal magnitude of compressive force. This study helps to define the underlying mechanism of compressive stimulation and may also enhance the application of compressive stimulation or CGRP peptide as an alternative approach for accelerating tooth movement in orthodontic treatment.
Collapse
Affiliation(s)
- Thunwa Binlateh
- School of Pharmacy, Walailak University, Nakhon Si Thammarat, Thailand
| | - Chidchanok Leethanakul
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
| | - Peungchaleoy Thammanichanon
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima, Thailand.
- Oral Health Center, Suranaree University of Technology Hospital, Suranaree University of Technology, Nakhon Ratchasima, Thailand.
| |
Collapse
|
7
|
Xiong L, Guo HH, Pan JX, Ren X, Lee D, Chen L, Mei L, Xiong WC. ATP6AP2, a regulator of LRP6/β-catenin protein trafficking, promotes Wnt/β-catenin signaling and bone formation in a cell type dependent manner. Bone Res 2024; 12:33. [PMID: 38811544 PMCID: PMC11137048 DOI: 10.1038/s41413-024-00335-7] [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: 10/11/2023] [Revised: 03/06/2024] [Accepted: 04/08/2024] [Indexed: 05/31/2024] Open
Abstract
Wnt/β-catenin signaling is critical for various cellular processes in multiple cell types, including osteoblast (OB) differentiation and function. Exactly how Wnt/β-catenin signaling is regulated in OBs remain elusive. ATP6AP2, an accessory subunit of V-ATPase, plays important roles in multiple cell types/organs and multiple signaling pathways. However, little is known whether and how ATP6AP2 in OBs regulates Wnt/β-catenin signaling and bone formation. Here we provide evidence for ATP6AP2 in the OB-lineage cells to promote OB-mediated bone formation and bone homeostasis selectively in the trabecular bone regions. Conditionally knocking out (CKO) ATP6AP2 in the OB-lineage cells (Atp6ap2Ocn-Cre) reduced trabecular, but not cortical, bone formation and bone mass. Proteomic and cellular biochemical studies revealed that LRP6 and N-cadherin were reduced in ATP6AP2-KO BMSCs and OBs, but not osteocytes. Additional in vitro and in vivo studies revealed impaired β-catenin signaling in ATP6AP2-KO BMSCs and OBs, but not osteocytes, under both basal and Wnt stimulated conditions, although LRP5 was decreased in ATP6AP2-KO osteocytes, but not BMSCs. Further cell biological studies uncovered that osteoblastic ATP6AP2 is not required for Wnt3a suppression of β-catenin phosphorylation, but necessary for LRP6/β-catenin and N-cadherin/β-catenin protein complex distribution at the cell membrane, thus preventing their degradation. Expression of active β-catenin diminished the OB differentiation deficit in ATP6AP2-KO BMSCs. Taken together, these results support the view for ATP6AP2 as a critical regulator of both LRP6 and N-cadherin protein trafficking and stability, and thus regulating β-catenin levels, demonstrating an un-recognized function of osteoblastic ATP6AP2 in promoting Wnt/LRP6/β-catenin signaling and trabecular bone formation.
Collapse
Affiliation(s)
- Lei Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA
| | - Hao-Han Guo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA
| | - Jin-Xiu Pan
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA
| | - Xiao Ren
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Daehoon Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA
| | - Li Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
8
|
Song X, Dong C, Man X. Phosphorylated MAPK11 promotes the progression of clear cell renal cell carcinoma by maintaining RUNX2 protein abundance. J Cell Mol Med 2023; 27:2583-2593. [PMID: 37525479 PMCID: PMC10468653 DOI: 10.1111/jcmm.17870] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 06/20/2023] [Accepted: 07/16/2023] [Indexed: 08/02/2023] Open
Abstract
Previous studies have demonstrated that mitogen-activated protein kinase 11 (MAPK11) functions as an important point of integration in signalling transduction pathways and controlling endocellular processes, including viability of cells, differentiation, proliferation and apoptosis, through the sequence phosphorylation of the substrate protein Ser/Thr kinase protein cascade. Though MAPK 11 plays an important role in various tumours, especially in the invasive and metastatic processes, its expression and molecular mechanism in clear cell renal cell carcinoma (ccRCC) remain unclear. Runt-associated transcription factor 2 (RUNX2), a main transcription factor for osteoblast differentiation and chondrocyte maturation, has high expression in a number of tumours. In this study, the mRNA and protein levels of targeted genes in ccRCC tissues and adjacent tissues are analysed using the Cancer Genome Atlas (TCGA) database and western blotting. The ccRCC cell proliferation was measured with colony formation and EdU assay, and cell migration was examined through transwell assay. The interactive behaviour between proteins was detected with immunoprecipitation. Half-life period of RUNX2 protein was measured with cycloheximide chase assay. The results of the study indicated overexpression of MAPK11 and RUNX2 in ccRCC tissues and cell lines. MAPK11 and RUNX2 promoted the ccRCC cell proliferation and migration. Additionally, physical interaction took place between RUNX2 and P-MAPK11, which functioned to sustain the stability of RUNX2 protein. The high expression of RUNX2 could neutralize the functional degradation in MAPK11. And the outcomes of the study suggest that the P-MAPK11/RUNX2 axis may be used as a potential therapeutic target of ccRCC.
Collapse
Affiliation(s)
- Xiandong Song
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Changming Dong
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xiaojun Man
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
9
|
Agriesti F, Landini F, Tamma M, Pacelli C, Mazzoccoli C, Calice G, Ruggieri V, Capitanio G, Mori G, Piccoli C, Capitanio N. Bioenergetic profile and redox tone modulate in vitro osteogenesis of human dental pulp stem cells: new perspectives for bone regeneration and repair. Stem Cell Res Ther 2023; 14:215. [PMID: 37608350 PMCID: PMC10463344 DOI: 10.1186/s13287-023-03447-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Redox signaling and energy metabolism are known to be involved in controlling the balance between self-renewal and proliferation/differentiation of stem cells. In this study we investigated metabolic and redox changes occurring during in vitro human dental pulp stem cells (hDPSCs) osteoblastic (OB) differentiation and tested on them the impact of the reactive oxygen species (ROS) signaling. METHODS hDPSCs were isolated from dental pulp and subjected to alkaline phosphatase and alizarin red staining, q-RT-PCR, and western blotting analysis of differentiation markers to assess achievement of osteogenic/odontogenic differentiation. Moreover, a combination of metabolic flux analysis and confocal cyto-imaging was used to profile the metabolic phenotype and to evaluate the redox tone of hDPSCs. RESULTS In differentiating hDPSCs we observed the down-regulation of the mitochondrial respiratory chain complexes expression since the early phase of the process, confirmed by metabolic flux analysis, and a reduction of the basal intracellular peroxide level in its later phase. In addition, dampened glycolysis was observed, thereby indicating a lower energy-generating phenotype in differentiating hDPSCs. Treatment with the ROS scavenger Trolox, applied in the early-middle phases of the process, markedly delayed OB differentiation of hDPSCs assessed as ALP activity, Runx2 expression, mineralization capacity, expression of stemness and osteoblast marker genes (Nanog, Lin28, Dspp, Ocn) and activation of ERK1/2. In addition, the antioxidant partly prevented the inhibitory effect on cell metabolism observed following osteogenic induction. CONCLUSIONS Altogether these results provided evidence that redox signaling, likely mediated by peroxide species, influenced the stepwise osteogenic expansion/differentiation of hDPSCs and contributed to shape its accompanying metabolic phenotype changes thus improving their efficiency in bone regeneration and repair.
Collapse
Affiliation(s)
- Francesca Agriesti
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Francesca Landini
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Mirko Tamma
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Carmela Mazzoccoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Giovanni Calice
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Vitalba Ruggieri
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
- Clinical Pathology Unit, “Madonna delle Grazie’’ Hospital, Matera, Italy
| | - Giuseppe Capitanio
- Department of Translational Biomedicine and Neuroscience “DiBraiN”, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
10
|
Yun H, Kim B, Jeong YH, Hong JT, Park K. Suffruticosol A elevates osteoblast differentiation targeting BMP2-Smad/1/5/8-RUNX2 in pre-osteoblasts. Biofactors 2023; 49:127-139. [PMID: 35852295 PMCID: PMC10947220 DOI: 10.1002/biof.1878] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/03/2022] [Indexed: 11/11/2022]
Abstract
The Paeonia suffruticosa ANDR. (P. suffruticosa) is commonly used in traditional medicine for various purposes. Suffruticosol A (Suf-A), isolated from P. suffruticosa, is a beneficial compound with antibiofilm, antivirulence, and anti-inflammatory properties. The aim of the present study was to investigate the biological effects of Suf-A on osteogenic processes in pre-osteoblasts. It was determined here in that Suf-A (>98.02%), isolated from P. suffruticosa, showed no cytotoxicity at 0.1-30 μM; however, it induced cytotoxicity at 50-100 μM in pre-osteoblasts. Suf-A increased osteogenic alkaline phosphatase activity and expression levels of noncollagenous proteins. Adhesion and trans-migration on the extracellular matrix were potentiated by Suf-A, but not by wound-healing migration. Suf-A did not affect autophagy or necroptosis during osteoblast differentiation. We found that Suf-A increased runt-related transcription factor 2 (RUNX2) levels and mineralized matrix formation. RUNX2 expression was mediated by Suf-A-induced BMP2-Smad1/5/8 and mitogen-activated protein kinase signaling, as demonstrated by Noggin, a BMP2 inhibitor. These results suggest that Suf-A is a potential natural osteogenic compound.
Collapse
Affiliation(s)
- Hyung‐Mun Yun
- Department of Oral and Maxillofacial PathologySchool of Dentistry, Kyung Hee UniversitySeoulRepublic of Korea
| | - Bomi Kim
- National Development Institute of Korean MedicineGyeongsanRepublic of Korea
| | - Yun Hee Jeong
- National Development Institute of Korean MedicineGyeongsanRepublic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National UniversityChungbukRepublic of Korea
| | - Kyung‐Ran Park
- Gwangju CenterKorea Basic Science Institute (KBSI)GwangjuRepublic of Korea
| |
Collapse
|
11
|
Synergistic effects of enamel matrix derivatives and surface morphology of anodized titanium on osteogenic differentiation of bone marrow mesenchymal stem cells sheet. Am J Transl Res 2023; 15:1085-1096. [PMID: 36915734 PMCID: PMC10006779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/29/2022] [Indexed: 03/16/2023]
Abstract
OBJECTIVE To explore the potential osteogenic induction mechanism of enamel matrix derivatives (EMDs) on bone marrow mesenchymal stem cell (BMSC) sheets with different titanium surface morphologies. METHODS The BMSCs were inoculated on the surfaces of titanium alloys with different morphologies: anodic oxidation (AO), sand-blasted, large grit and acid-etched, and no treatment (control). The proliferation and osteogenic differentiation of BMSCs on the different surface morphologies were observed with the same concentration of EMDs. To further understand the osteogenic mechanism of EMDs on BMSC sheets with different morphologies, a real-time RT-PCR and a western blot were used to detect the overall levels of osteogenic genes and osteogenic proteins. Finally, to verify the osteogenic effect of BMSC sheets stimulated by EMDs in vivo, BMSC sheets with different morphologies were implanted into the subcutaneous tissue of the back of nude mice, and the bone formation was detected by HE staining. RESULTS The EMDs and surface morphology in the AO group synergically increased the expression levels of osteogenic active factors (RUNX2, OSX and OCN) and enhanced the osteogenic differentiation effect of BMSCs. The in vivo experiments showed that the BMSC sheets in the AO group were rich in osteogenic active factors, and promoted the formation of ectopic bone tissue after implantation into the subcutaneous tissue of the back of nude mice. CONCLUSION EMDs and AO morphology synergically enhance the secretion of bone osteogenic active factors of BMSCs and promote the formation of heterotopic bone.
Collapse
|
12
|
Gholami L, Afshar S, Arkian A, Saeidijam M, Hendi SS, Mahmoudi R, Khorsandi K, Hashemzehi H, Fekrazad R. NIR irradiation of human buccal fat pad adipose stem cells and its effect on TRP ion channels. Lasers Med Sci 2022; 37:3681-3692. [PMID: 36227520 DOI: 10.1007/s10103-022-03652-7] [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: 09/06/2021] [Accepted: 10/02/2022] [Indexed: 11/28/2022]
Abstract
The effect of near infrared (NIR) laser irradiation on proliferation and osteogenic differentiation of buccal fat pad-derived stem cells and the role of transient receptor potential (TRP) channels was investigated in the current research. After stem cell isolation, a 940 nm laser with 0.1 W, 3 J/cm2 was used in pulsed and continuous mode for irradiation in 3 sessions once every 48 h. The cells were cultured in the following groups: non-osteogenic differentiation medium/primary medium (PM) and osteogenic medium (OM) groups with laser-irradiated (L +), without irradiation (L -), laser treated + Capsazepine inhibitor (L + Cap), and laser treated + Skf96365 inhibitor (L + Skf). Alizarin Red staining and RT-PCR were used to assess osteogenic differentiation and evaluate RUNX2, Osterix, and ALP gene expression levels. The pulsed setting showed the best viability results (P < 0.05) and was used for osteogenic differentiation evaluations. The results of Alizarin red staining were not statistically different between the four groups. Osterix and ALP expression increased in the (L +) group. This upregulation abrogated in the presence of Capsazepine, TRPV1 inhibitor (L + Cap); however, no significant effect was observed with Skf96365 (L + Skf).
Collapse
Affiliation(s)
- Leila Gholami
- Department of Periodontics, Dental Implants Research Center, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saeid Afshar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Aliasghar Arkian
- Dental Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masood Saeidijam
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyedeh Sareh Hendi
- Department of Endodontics, Dental Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roghayeh Mahmoudi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Khatereh Khorsandi
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran.,Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Hadi Hashemzehi
- Department of Oral and Maxillofacial Surgery, Dental Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Reza Fekrazad
- Radiation Sciences Research Center, Laser Research Center in Medical Sciences, AJA University of Medical Sciences, Tehran, Iran. .,International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
13
|
Li Z, Wu Z, Xi X, Zhao F, Liu H, Liu D. Cellular communication network factor 1 interlinks autophagy and ERK signaling to promote osteogenesis of periodontal ligament stem cells. J Periodontal Res 2022; 57:1169-1182. [PMID: 36199215 DOI: 10.1111/jre.13054] [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: 06/07/2022] [Revised: 08/23/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To investigate the effects of cellular communication network factor 1 (CCN1), a critical matricellular protein, on alveolar bone regeneration, and to elucidate the underlying molecular mechanism. BACKGROUND In the process of orthodontic tooth movement, bone deposition on the tension side of human periodontal ligament stem cells (hPDLSCs) ensured high efficiency and long-term stability of the treatment. The matricellular protein CCN1 is responsive to mechanical stimulation, exhibiting important tasks in bone homoeostasis. However, the role and mechanism of CCN1 on alveolar bone remodeling of hPDLSCs remains unclear. METHODS The expression and distribution of CCN1 in rat periodontal ligament were detected by immunofluorescence staining and immunohistochemical staining. ELISA verified the secretion of CCN1 triggered by stretch loading. To examine the mineralization ability of hPDLSCs induced by CCN1, Western blotting, qRT-PCR, ARS, and ALP staining were performed. CCK-8 and cell migration assay were performed to detect the cell proliferation rate and the wound healing. PI3K/Akt, MAPK, and autophagy activation were examined via Western blotting and immunofluorescence. RESULTS Mechanical stimuli induced the release of CCN1 into extracellular environment by hPDLSCs. Knockdown of CCN1 attenuated the osteogenesis of hPDLSCs while rhCCN1 enhanced the expression of Runx2, Col 1, ALPL, and promoted the mineralization nodule formation. CCN1 activated PI3K/Akt and ERK signaling, and blockage of PI3K/Akt signaling reversed the accelerated cell migration triggered by CCN1. The enhanced osteogenesis induced by CCN1 was abolished by ERK signaling inhibitor PD98059 or autophagy inhibitor 3-MA. Further investigation demonstrated PD98059 abrogated the activation of autophagy. CONCLUSION This study demonstrated that CCN1 promotes osteogenesis in hPDLSCs via autophagy and MAPK/ERK pathway.
Collapse
Affiliation(s)
- Zixuan Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Zuping Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Xun Xi
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Fang Zhao
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Hong Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Dongxu Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
14
|
Kim H, Oh N, Kwon M, Kwon OH, Ku S, Seo J, Roh S. Exopolysaccharide of Enterococcus faecium L15 promotes the osteogenic differentiation of human dental pulp stem cells via p38 MAPK pathway. Stem Cell Res Ther 2022; 13:446. [PMID: 36056447 PMCID: PMC9440579 DOI: 10.1186/s13287-022-03151-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Background Bone has important functions in the body. Several researchers have reported that the polysaccharides and lipopolysaccharide derived from microbes can promote osteogenic differentiation of stem cells. Enterococcus faecium, a lactic acid bacterium (LAB), produces several bioactive metabolites and has been widely applied in the food and nutraceutical industries. The exopolysaccharide (EPS) from LAB has also been extensively examined for its postbiotic effects and for its in vivo and in vitro functionalities. However, studies on promoting bone differentiation using polysaccharides from LAB are lacking. Therefore, the purpose of this study was to investigate the effect of E. faecium L15 extract and EPS on osteogenic differentiation of human dental pulp stem cells (hDPSCs) and to identify the underlying mechanisms. Methods hDPSCs were obtained from dental pulp tissue, and L15 extract and EPS were isolated from L15. Gene and protein expression of the osteogenic differentiation markers were analyzed with qPCR and western blotting and the possible signaling pathways were also investigated using western blotting. Osteogenic differentiation potential was examined by alkaline phosphatase (ALP) staining and alizarin red s (ARS) staining. In addition, osteogenic differentiation potential of L15 EPS was explored in ex vivo culture of neonate murine calvaria. Results The calcium deposition and ALP activity were enhanced by addition of L15 extract or EPS. The expression levels of RUNX2, ALP, and COL1A1 mRNA and the protein expression levels of RUNX2, ALP, and BMP4 were increased in hDPSCs treated with the L15 extract or EPS. The L15 EPS treatment enhanced phosphorylation of the p38 mitogen-activated protein kinase (MAPK). The L15 EPS-induced increases in RUNX2, ALP, and BMP4 expression were suppressed by the p38 MAPK inhibitor SB203580. The promoting effect of L15 EPS on osteogenic differentiation was not only seen in hDPSCs, but also in osteoblast precursors. ALP activity and the expression of RUNX2, ALP, and COL1A1 increased in the L15 EPS-treated osteoblast precursors. In addition, L15 EPS increased bone thickness of neonate murine calvaria in ex vivo culture. Conclusions The stimulatory effect of L15 extract and EPS on osteogenic differentiation occurred through the p38 MAPK pathway, and L15 EPS enhanced new bone formation in neonate murine calvaria. These data suggest that L15 EPS has therapeutic potential applicable to bone regeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03151-0.
Collapse
Affiliation(s)
- Hyewon Kim
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 08826, Korea
| | - Naeun Oh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 08826, Korea
| | - Mijin Kwon
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 08826, Korea
| | - Oh-Hee Kwon
- Biomedical Research Institute, NeoRegen Biotech Co., Ltd., Gyeonggi-do, 16641, Korea
| | - Seockmo Ku
- Fermentation Science Program, School of Agriculture, College of Basic and Applied Sciences, Middle Tennessee State University, Murfreesboro, TN, 37132, USA
| | - Jeongmin Seo
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 08826, Korea. .,Biomedical Research Institute, NeoRegen Biotech Co., Ltd., Gyeonggi-do, 16641, Korea.
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 08826, Korea.
| |
Collapse
|
15
|
A Low-Phenylalanine-Containing Whey Protein Hydrolysate Stimulates Osteogenic Activity through the Activation of p38/Runx2 Signaling in Osteoblast Cells. Nutrients 2022; 14:nu14153135. [PMID: 35956311 PMCID: PMC9370741 DOI: 10.3390/nu14153135] [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: 06/27/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/04/2022] Open
Abstract
A phenylalanine (Phe)-restricted diet is indispensable for individuals suffering from phenylketonuria (PKU). Our previous study reported a low-Phe-containing whey protein hydrolysate (LPH) prepared from a selected whey protein hydrolysate (TA2H). This study aimed to investigate the osteogenic activity of LPH and TA2H in MC3T3-E1 preosteoblast cells and explore the underlying mechanism. Results showed that the treatment of TA2H and LPH (at the final concentrations of 100–1000 μg/mL) had a stimulatory effect on the proliferation, differentiation, and mineralization of MC3T3-E1 cells. The LPH of 1000 μg/mL significantly increased cell proliferation (2.15- ± 0.11-fold) and alkaline phosphatase activity (1.22- ± 0.07-fold), promoted the protein and mRNA levels of runt-related transcription factor 2 (Runx2, 2.50- ± 0.14-fold and 2.97- ± 0.23-fold, respectively), enhanced the expression of differentiation biomarkers (type-I collagen, osteocalcin, and osteopontin), increased calcium deposition (1.56- ± 0.08-fold), and upregulated the ratio of osteoprotegerin/receptor activator of nuclear factor-κB ligand. The exploration of signaling pathways indicated that the activated p38-dependent Runx2 signaling contributed to the LPH-induced osteogenesis. These results provided evidence, for the first time, that a prepared low-Phe whey protein hydrolysate positively modulated the activity of osteoblasts through the p38/Runx2 pathway, thereby providing a new osteoinductive protein substitute to make functional PKU food.
Collapse
|
16
|
Park KR, Kim B, Lee JY, Moon HJ, Kwon IK, Yun HM. Effects of Scoparone on differentiation, adhesion, migration, autophagy and mineralization through the osteogenic signalling pathways. J Cell Mol Med 2022; 26:4520-4529. [PMID: 35796406 PMCID: PMC9357629 DOI: 10.1111/jcmm.17476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/30/2022] [Accepted: 06/15/2022] [Indexed: 12/18/2022] Open
Abstract
Scoparone (SCOP), an active and efficient coumarin compound derived from Artemisia capillaris Thunb, has been used as a traditional Chinese herbal medicine. Herein, we investigated the effects of SCOP on the osteogenic processes using MC3T3‐E1 pre‐osteoblasts in in vitro cell systems. SCOP (C11H10O4, > 99.17%) was purified and identified from A. capillaries. SCOP (0.1 to 100 μM concentrations) did not have cytotoxic effects in pre‐osteoblasts; however, it promoted alkaline phosphatase (ALP) staining and activity, and mineralized nodule formation under early and late osteogenic induction. SCOP elevated osteogenic signals through the bone morphogenetic protein 2 (BMP2)‐Smad1/5/8 pathway, leading to the increased expression of runt‐related transcription factor 2 (RUNX2) with its target protein, matrix metallopeptidase 13 (MMP13). SCOP also induced the non‐canonical BMP2‐MAPKs pathway, but not the Wnt3a‐β‐catenin pathway. Moreover, SCOP promoted autophagy, migration and adhesion under the osteogenic induction. Overall, the findings of this study demonstrated that SCOP has osteogenic effects associated with cell differentiation, adhesion, migration, autophagy and mineralization.
Collapse
Affiliation(s)
- Kyung-Ran Park
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, Korea
| | - Bomi Kim
- National Development Institute of Korean Medicine, Gyeongsan, Korea
| | - Joon Yeop Lee
- National Development Institute of Korean Medicine, Gyeongsan, Korea
| | - Ho-Jin Moon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Korea.,Medical Device Research Center, Medical Science Research Institute, Kyung Hee University Medical Center, Seoul, Korea
| | - Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul, Korea
| |
Collapse
|
17
|
Marsh S, Constantin-Teodosiu T, Chapman V, Sottile V. In vitro Exposure to Inflammatory Mediators Affects the Differentiation of Mesenchymal Progenitors. Front Bioeng Biotechnol 2022; 10:908507. [PMID: 35813997 PMCID: PMC9257013 DOI: 10.3389/fbioe.2022.908507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/23/2022] [Indexed: 11/27/2022] Open
Abstract
The increasing prevalence of joint disease, and in particular osteoarthritis (OA), calls for novel treatment strategies to prevent disease progression in addition to existing approaches focusing mainly on the relief of pain symptoms. The inherent properties of mesenchymal stem cells (MSCs) make them an attractive candidate for novel tissue repair strategies, as these progenitors have the potential to differentiate into chondrocytes needed to replace degraded cartilage and can exert a modulating effect on the inflammatory environment of the diseased joint. However, the inflammatory environment of the joint may affect the ability of these cells to functionally integrate into the host tissue and exert beneficial effects, as hinted by a lack of success seen in clinical trials. Identification of factors and cell signalling pathways that influence MSC function is therefore critical for ensuring their success in the clinic, and here the effects of inflammatory mediators on bone marrow-derived MSCs were evaluated. Human MSCs were cultured in the presence of inflammatory mediators typically associated with OA pathology (IL-1β, IL-8, IL-10). While exposure to these factors did not produce marked effects on MSC proliferation, changes were observed when the mediators were added under differentiating conditions. Results collected over 21 days showed that exposure to IL-1β significantly affected the differentiation response of these cells exposed to chondrogenic and osteogenic conditions, with gene expression analysis indicating changes in MAPK, Wnt and TLR signalling pathways, alongside an increased expression of pro-inflammatory cytokines and cartilage degrading enzymes. These results highlight the value of MSCs as a preclinical model to study OA and provide a basis to define the impact of factors driving OA pathology on the therapeutic potential of MSCs for novel OA treatments.
Collapse
Affiliation(s)
- S. Marsh
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Pain Centre Versus Arthritis, University of Nottingham, Nottingham, United Kingdom
| | - T. Constantin-Teodosiu
- Pain Centre Versus Arthritis, University of Nottingham, Nottingham, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - V. Chapman
- Pain Centre Versus Arthritis, University of Nottingham, Nottingham, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - V. Sottile
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Pain Centre Versus Arthritis, University of Nottingham, Nottingham, United Kingdom
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- *Correspondence: V. Sottile,
| |
Collapse
|
18
|
Nihashi Y, Miyoshi M, Umezawa K, Shimosato T, Takaya T. Identification of a Novel Osteogenetic Oligodeoxynucleotide (osteoDN) That Promotes Osteoblast Differentiation in a TLR9-Independent Manner. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1680. [PMID: 35630904 PMCID: PMC9145662 DOI: 10.3390/nano12101680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/06/2022] [Accepted: 05/13/2022] [Indexed: 12/11/2022]
Abstract
Dysfunction of bone-forming cells, osteoblasts, is one of the causes of osteoporosis. Accumulating evidence has indicated that oligodeoxynucleotides (ODNs) designed from genome sequences have the potential to regulate osteogenic cell fate. Such osteogenetic ODNs (osteoDNs) targeting and activating osteoblasts can be the candidates of nucleic acid drugs for osteoporosis. In this study, the ODN library derived from the Lacticaseibacillus rhamnosus GG genome was screened to determine its osteogenetic effect on murine osteoblast cell line MC3T3-E1. An 18-base ODN, iSN40, was identified to enhance alkaline phosphatase activity of osteoblasts within 48 h. iSN40 also induced the expression of osteogenic genes such as Msx2, osterix, collagen type 1α, osteopontin, and osteocalcin. Eventually, iSN40 facilitated calcium deposition on osteoblasts at the late stage of differentiation. Intriguingly, the CpG motif within iSN40 was not required for its osteogenetic activity, indicating that iSN40 functions in a TLR9-independent manner. These data demonstrate that iSN40 serves as a novel osteogenetic ODN (osteoDN) that promotes osteoblast differentiation. iSN40 provides a potential seed of the nucleic acid drug that activating osteoblasts for osteoporosis therapy.
Collapse
Affiliation(s)
- Yuma Nihashi
- Department of Science and Technology, Graduate School of Medicine, Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (Y.N.); (T.S.)
| | - Mana Miyoshi
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
| | - Koji Umezawa
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Takeshi Shimosato
- Department of Science and Technology, Graduate School of Medicine, Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (Y.N.); (T.S.)
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Tomohide Takaya
- Department of Science and Technology, Graduate School of Medicine, Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (Y.N.); (T.S.)
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan;
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| |
Collapse
|
19
|
Wang B, Gong S, Han L, Shao W, Li Z, Xu J, Lv X, Xiao B, Feng Y. Knockdown of HDAC9 Inhibits Osteogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells Partially by Suppressing the MAPK Signaling Pathway. Clin Interv Aging 2022; 17:777-787. [PMID: 35592642 PMCID: PMC9113040 DOI: 10.2147/cia.s361008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/05/2022] [Indexed: 11/23/2022] Open
Abstract
Background Histone deacetylase 9 (HDAC9) is a member of the HDAC gene family that plays essential roles in the organization of transcriptional regulation by catalyzing deacetylation of histone proteins. However, the effects of HDAC9 on osteonecrosis of femoral head (ONFH) have not been investigated. The present study aimed to reveal whether histone deacetylase 9 (HDAC9) regulated osteogenic differentiation. Methods A lentiviral knockdown HDAC9 model was established in hBMSCs. Osteoblast-specific gene expression, such as Runx2, OCN was examined by qRT-PCR and Western blot, respectively. Though transcriptome sequencing and enrichment analysis, related signal pathways caused by down-regulation of HDAC9 were screened. The effect of HDAC9 on MAPK signaling pathway was determined by Western blot. Eventually, tert-Butylhydroquinone (tBHQ) was used to examine the effect of MAPK activation on osteogenesis in HDAC9 knockdown hBMSCs. Results A lentiviral knockdown HDAC9 model was successfully established in hBMSCs. HDAC9 knockdown significantly inhibited osteoblast-specific gene expression, such as runt-related transcription factor 2 (Runx2), osteocalcin (OCN) and mineral deposition in vitro. Moreover, a total of 950 DEGs were identified in HDAC9-knockdown hBMSCs. We discovered that the MAPK signaling pathway might be related to this process by pathway enrichment analysis. HDAC9 knockdown significantly reduced the expression level of phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2). Finally, the decreased osteogenesis due to HDAC9 knockdown was partly rescued by a MAPK signaling pathway activator. Conclusion Taken together, these results suggest that HDAC9 knockdown inhibits osteogenic differentiation of hBMSCs, partially through the MAPK signaling pathway. HDAC9 may serve as a potential target for the treatment of ONFH.
Collapse
Affiliation(s)
- Bo Wang
- Department of Rehabilitation, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Song Gong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Lizhi Han
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Wenkai Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Zilin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Jiawei Xu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Xiao Lv
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Baojun Xiao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Correspondence: Baojun Xiao; Yong Feng, Email ;
| | - Yong Feng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| |
Collapse
|
20
|
Yadav LR, Balagangadharan K, Lavanya K, Selvamurugan N. Orsellinic acid-loaded chitosan nanoparticles in gelatin/nanohydroxyapatite scaffolds for bone formation in vitro. Life Sci 2022; 299:120559. [PMID: 35447131 DOI: 10.1016/j.lfs.2022.120559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
AIM Orsellinic acid (2,4-Dimethoxy-6-methylbenzoic acid) (OA) is a hydrophobic polyphenolic compound with therapeutic potential, but its impact on actuating osteogenesis remains unknown. The bioavailability of OA is hampered by its hydrophobic nature. This study aimed to fabricate nano-drug delivery system-based scaffolds for OA and test its potential for osteogenesis in vitro. MATERIALS AND METHODS OA was loaded into chitosan nanoparticles (nCS + OA) using the ionic gelation technique at different concentrations. nCS + OA were incorporated onto the scaffolds containing gelatin (Gel) and nanohydroxyapatite (nHAp) by the lyophilization method. Biocomposite scaffolds were examined for their physicochemical and material characteristic properties. The effect of OA in the scaffolds for osteoblast differentiation was determined by alizarin red and von Kossa staining at the cellular level and by reverse transcriptase-qPCR and western blot analysis at the molecular level. KEY FINDINGS The scaffolds showed excellent physiochemical and material characteristics and remained cyto-friendly to mouse mesenchymal stem cells (mMSCs, C3H10T1/2). The release of OA from Gel/nHAp/nCS scaffolds enhanced the differentiation of mMSCs towards osteoblasts, as observed through cellular and molecular studies. Moreover, the osteogenic potential of OA was mediated by the activation of FAK and ERK signaling pathways through integrins. SIGNIFICANCE The inclusion of OA into Gel/nHAp/nCS biocomposite scaffolds at 80 μM concentration promoted osteoblast differentiation via cell adhesion mediated signaling, compared with that shown by Gel/nHAp/nCS alone. Overall, this study identified the potential therapeutic OA containing Gel/nHAp/nCS scaffolds, accelerating its potential for clinical application towards bone regeneration.
Collapse
Affiliation(s)
- L Roshini Yadav
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - K Balagangadharan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - K Lavanya
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
21
|
Bu T, Zhang L, Liu L, Yu S, Zheng J, Wu J, Yang K. Evaluation of the anti-osteoporotic effect of a low-phenylalanine whey protein hydrolysate in an ovariectomized mice model. Food Funct 2022; 13:3957-3967. [PMID: 35293905 DOI: 10.1039/d1fo04030h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A phenylalanine (Phe)-restricted diet is indispensable to control the blood Phe for individuals with phenylketonuria (PKU), who are also confronted with progressive bone impairment. Thus, the development of a low-Phe protein substitute that could positively regulate bone metabolism is desired for their bone health. Our previous study reported the preparation of a low-Phe containing whey hydrolysate (LPH) from a selected whey protein hydrolysate (TAH). However, the effect of LPH on the bone status is unknown. In this study, we used an ovariectomized (OVX) mice model to evaluate the anti-osteoporotic potential of oral administration of whey protein concentrate (WPC, protein control), TAH, and LPH on bone physiology and bone metabolism. The results showed that after 12 weeks of treatment, the decreased bone mineral density, the deteriorated trabecular microarchitecture, and the reduced ultimate load due to ovariectomy were significantly attenuated by two whey protein hydrolysates (TAH and LPH); meanwhile, the body weight, uterine weight, bone composition, and the femoral elastic load of OVX mice had not been significantly affected by whey samples. In addition, LPH and TAH dual-regulated bone remodeling in OVX mice through triggering osteogenesis (promoted the expression of runt-related protein 2 (Runx2) and osteoformation markers) and inhibiting osteoresorption as well as inflammation. The modulated mitogen-activated protein kinase signaling and the inhibited nuclear factor κB signaling by LPH and TAH might relate to the dual-regulatory activities on bone. Overall, in the OVX mice model, LPH exerted higher osteoprotective potential than TAH of the same dose by activating the bone formation markers and inhibiting the inflammatory status. The current study demonstrated for the first time the potential use of a low-Phe whey hydrolysate, a protein substitute for PKU individuals, in the prevention of osteoporosis.
Collapse
Affiliation(s)
- Tingting Bu
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Ling Zhang
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Ling Liu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Songfeng Yu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jiexia Zheng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, T6G 2P5, Canada
| | - Kai Yang
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
22
|
Gao L, Zhang SQ. Antiosteoporosis Effects, Pharmacokinetics, and Drug Delivery Systems of Icaritin: Advances and Prospects. Pharmaceuticals (Basel) 2022; 15:397. [PMID: 35455393 PMCID: PMC9032325 DOI: 10.3390/ph15040397] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/10/2022] [Accepted: 03/22/2022] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis is a systemic skeletal disorder affecting over 200 million people worldwide and contributes dramatically to global healthcare costs. Available anti-osteoporotic drug treatments including hormone replacement therapy, anabolic agents, and bisphosphonates often cause adverse events which limit their long-term use. Therefore, the application of natural products has been proposed as an alternative therapy strategy. Icaritin (ICT) is not only an enzyme-hydrolyzed product of icariin but also an intestinal metabolite of eight major flavonoids of the traditional Chinese medicinal plant Epimedium with extensive pharmacological activities, such as strengthening the kidney and reinforcing the bone. ICT displays several therapeutic effects, including osteoporosis prevention, neuroprotection, antitumor, cardiovascular protection, anti-inflammation, and immune-protective effect. ICT inhibits bone resorption activity of osteoclasts and stimulates osteogenic differentiation and maturation of bone marrow stromal progenitor cells and osteoblasts. As for the mechanisms of effect, ICT regulates relative activities of two transcription factors Runx2 and PPARγ, determines the differentiation of MSCs into osteoblasts, increases mRNA expression of OPG, and inhibits mRNA expression of RANKL. Poor water solubility, high lipophilicity, and unfavorable pharmacokinetic properties of ICT restrict its anti-osteoporotic effects, and novel drug delivery systems are explored to overcome intrinsic limitations of ICT. The paper focuses on osteogenic effects and mechanisms, pharmacokinetics and delivery systems of ICT, and highlights bone-targeting strategies to concentrate ICT on the ideal specific site of bone. ICT is a promising potential novel therapeutic agent for osteoporosis.
Collapse
Affiliation(s)
- Lifang Gao
- School of Public Health, Capital Medical University, 10 Youanmenwai Xitiao, Beijing 100069, China;
| | - Shuang-Qing Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Beijing 100050, China
| |
Collapse
|
23
|
Gomez GA, Aghajanian P, Pourteymoor S, Larkin D, Mohan S. Differences in pathways contributing to thyroid hormone effects on postnatal cartilage calcification versus secondary ossification center development. eLife 2022; 11:76730. [PMID: 35098920 PMCID: PMC8830887 DOI: 10.7554/elife.76730] [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: 12/30/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
The proximal and distal femur epiphyses of mice are both weight-bearing structures derived from chondrocytes but differ in development. Mineralization at the distal epiphysis occurs in an osteoblast-rich secondary ossification center (SOC), while the chondrocytes of the proximal femur head (FH), in particular, are directly mineralized. Thyroid hormone (TH) plays important roles in distal knee SOC formation, but whether TH also affects proximal FH development remains unexplored. Here, we found that TH controls chondrocyte maturation and mineralization at the FH in vivo through studies in thyroid stimulating hormone receptor (Tshr-/-) hypothyroid mice by X-ray, histology, transcriptional profiling, and immunofluorescence staining. Both in vivo and in vitro studies conducted in ATDC5 chondrocyte progenitors concur that TH regulates expression of genes that modulate mineralization (Ibsp, Bglap2, Dmp1, Spp1, and Alpl). Our work also delineates differences in prominent transcription factor regulation of genes involved in the different mechanisms leading to proximal FH cartilage calcification and endochondral ossification at the distal femur. The information on the molecular pathways contributing to postnatal cartilage calcification can provide insights on therapeutic strategies to treat pathological calcification that occurs in soft tissues such as aorta, kidney, and articular cartilage.
Collapse
Affiliation(s)
- Gustavo A Gomez
- Musculoskeletal Disease Centre, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, United States
| | | | - Sheila Pourteymoor
- Musculoskeletal Disease Centre, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, United States
| | - Destiney Larkin
- Musculoskeletal Disease Centre, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, United States
| | - Subburaman Mohan
- Musculoskeletal Disease Centre, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, United States
| |
Collapse
|
24
|
Abstract
Bone regeneration is a central focus of maxillofacial research, especially when dealing with dental implants or critical sized wound sites. While bone has great regeneration potential, exogenous delivery of growth factors can greatly enhance the speed, duration, and quality of osseointegration, making a difference in a patient’s quality of life. Bone morphogenic protein 2 (BMP-2) is a highly potent growth factor that acts as a recruiting molecule for mesenchymal stromal cells, induces a rapid differentiation of them into osteoblasts, while also maintaining their viability. Currently, the literature data shows that the liposomal direct delivery or transfection of plasmids containing BMP-2 at the bone wound site often results in the overexpression of osteogenic markers and result in enhanced mineralization with formation of new bone matrix. We reviewed the literature on the scientific data regarding BMP-2 delivery with the help of liposomes. This may provide the ground for a future new bone regeneration strategy with real chances of reaching clinical practice.
Collapse
|
25
|
Stimulation of the Runx2 P1 promoter by collagen-derived dipeptide prolyl-hydroxyproline bound to Foxg1 and Foxo1 in osteoblasts. Biosci Rep 2021; 41:230239. [PMID: 34779485 PMCID: PMC8655505 DOI: 10.1042/bsr20210304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 10/25/2021] [Accepted: 11/10/2021] [Indexed: 11/17/2022] Open
Abstract
Collagen-derived dipeptide prolyl-hydroxyproline (Pro-Hyp) directly binds to the forkhead box g1 (Foxg1) protein and causes it to undergo structural alteration. Pro-Hyp also promotes the production of a regulator of osteoblast differentiation, Runt-related transcription factor 2 (Runx2), through Foxg1, inducing osteoblast differentiation. In addition, Pro-Hyp disrupts the interaction between Foxg1 and Runx2, and Foxg1 appears to interact with Runx2 in the absence of Pro-Hyp. To elucidate the mechanism of Pro-Hyp that promotes osteoblast differentiation, we investigated whether Pro-Hyp regulates the Runx2 P1 promoter together with Foxg1. The present study revealed that Pro-Hyp is taken up by osteoblastic cells via the solute carrier family 15 member (Slc15a) 4. In the presence of Pro-Hyp, Runx2 is translocated from the nucleus to the cytoplasm and Foxg1 is translocated from the cytoplasm to the nucleus. We also found that Pro-Hyp promoted the interaction between Forkhead box o1 (Foxo1) and Runx2 and the dissociation of Foxg1 from Runx2. Moreover, we identified the Pro-Hyp response element in the Runx2 distal P1 promoter at nt −375 to −316, including the Runx2 binding sites and Fox core sequence. In the presence of Pro-Hyp, Runx2 is dissociated from the Pro-Hyp response element in the Runx2 distal P1 promoter. Subsequently, Foxg1 and Foxo1 activated the Runx2 promoter by binding to the Pro-Hyp response element. In summary, we delineated the mechanism by which Pro-Hyp stimulates the bone-related Runx2 distal P1 promoter activity in osteoblastic cells through Foxg1, Foxo1, and Runx2.
Collapse
|
26
|
Ohba S. Genome-scale actions of master regulators directing skeletal development. JAPANESE DENTAL SCIENCE REVIEW 2021; 57:217-223. [PMID: 34745394 PMCID: PMC8556520 DOI: 10.1016/j.jdsr.2021.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/14/2021] [Accepted: 10/10/2021] [Indexed: 11/03/2022] Open
Abstract
The mammalian skeleton develops through two distinct modes of ossification: intramembranous ossification and endochondral ossification. During the process of skeletal development, SRY-box containing gene 9 (Sox9), runt-related transcription factor 2 (Runx2), and Sp7 work as master transcription factors (TFs) or transcriptional regulators, underlying cell fate specification of the two distinct populations: bone-forming osteoblasts and cartilage-forming chondrocytes. In the past two decades, core transcriptional circuits underlying skeletal development have been identified mainly through mouse genetics and biochemical approaches. Recently emerging next-generation sequencer (NGS)-based studies have provided genome-scale views on the gene regulatory landscape programmed by the master TFs/transcriptional regulators. With particular focus on Sox9, Runx2, and Sp7, this review aims to discuss the gene regulatory landscape in skeletal development, which has been identified by genome-scale data, and provide future perspectives in this field.
Collapse
Affiliation(s)
- Shinsuke Ohba
- Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
27
|
Echave M, Erezuma I, Golafshan N, Castilho M, Kadumudi F, Pimenta-Lopes C, Ventura F, Pujol A, Jimenez J, Camara J, Hernáez-Moya R, Iturriaga L, Sáenz Del Burgo L, Iloro I, Azkargorta M, Elortza F, Lakshminarayanan R, Al-Tel T, García-García P, Reyes R, Delgado A, Évora C, Pedraz J, Dolatshahi-Pirouz A, Orive G. Bioinspired gelatin/bioceramic composites loaded with bone morphogenetic protein-2 (BMP-2) promote osteoporotic bone repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112539. [DOI: 10.1016/j.msec.2021.112539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 12/17/2022]
|
28
|
Park KR, Park JE, Kim B, Kwon IK, Hong JT, Yun HM. Calycosin-7-O-β-Glucoside Isolated from Astragalus membranaceus Promotes Osteogenesis and Mineralization in Human Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms222111362. [PMID: 34768792 PMCID: PMC8583672 DOI: 10.3390/ijms222111362] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/21/2022] Open
Abstract
Stem cells have received attention in various diseases, such as inflammatory, cancer, and bone diseases. Mesenchymal stem cells (MSCs) are multipotent stem cells that are critical for forming and repairing bone tissues. Herein, we isolated calycosin-7-O-β-glucoside (Caly) from the roots of Astragalus membranaceus, which is one of the most famous medicinal herbs, and investigated the osteogenic activities of Caly in MSCs. Caly did not affect cytotoxicity against MSCs, whereas Caly enhanced cell migration during the osteogenesis of MSCs. Caly increased the expression and enzymatic activities of ALP and the formation of mineralized nodules during the osteogenesis of MSCs. The osteogenesis and bone-forming activities of Caly are mediated by bone morphogenetic protein 2 (BMP2), phospho-Smad1/5/8, Wnt3a, phospho-GSK3β, and phospho-AKT, inducing the expression of runt-related transcription factor 2 (RUNX2). In addition, Caly-mediated osteogenesis and RUNX2 expression were attenuated by noggin and wortmannin. Moreover, the effects were validated in pre-osteoblasts committed to the osteoblast lineages from MSCs. Overall, our results provide novel evidence that Caly stimulates osteoblast lineage commitment of MSCs by triggering RUNX2 expression, suggesting Caly as a potential anabolic drug to prevent bone diseases.
Collapse
Affiliation(s)
- Kyung-Ran Park
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Korea;
- Medical Device Research Center, Medical Science Research Institute, Kyung Hee University Medical Center, Seoul 02447, Korea;
| | - Ji Eun Park
- National Institute for Korean Medicine Development, Gyeongsan 38540, Korea; (J.E.P.); (B.K.)
| | - Bomi Kim
- National Institute for Korean Medicine Development, Gyeongsan 38540, Korea; (J.E.P.); (B.K.)
| | - Il Keun Kwon
- Medical Device Research Center, Medical Science Research Institute, Kyung Hee University Medical Center, Seoul 02447, Korea;
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si 28160, Korea
- Correspondence: (J.T.H.); (H.-M.Y.); Tel.: +82-02-961-0691 (H.-M.Y.); Fax: +82-02-960-1457 (H.-M.Y.)
| | - Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Korea;
- Correspondence: (J.T.H.); (H.-M.Y.); Tel.: +82-02-961-0691 (H.-M.Y.); Fax: +82-02-960-1457 (H.-M.Y.)
| |
Collapse
|
29
|
Cheng L, Li Y, Xia Q, Meng M, Ye Z, Tang Z, Feng H, Chen X, Chen H, Zeng X, Luo Y, Dong Q. Enamel matrix derivative (EMD) enhances the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Bioengineered 2021; 12:7033-7045. [PMID: 34587869 PMCID: PMC8806549 DOI: 10.1080/21655979.2021.1971504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
To investigate the EMD's capacity in BMSCs osteogenic differentiation. In vivo and in vitro, BMSCs were treated with EMD, scanning electron microscopy, and Alizarin Red staining were used to detect the changes in the osteogenic ability of BMSCs, and the proliferation ability of BMSCs was evaluated by CCK8. In addition, by adding xav939, a typical inhibitor of Wnt/β-catenin signaling pathway, the regulatory function of Wnt/β-catenin signaling was clarified. The results showed that EMD promote cell proliferation and 25 μg/ml EMD had the most significant effect. Cells inducing osteogenesis for 2 and 3 even 4 weeks, the cell staining is deeper in EMD treated group than that of the control (P < 0.05) by alizarin Red staining, suggesting more mineralization of BMSCs. In vivo implanting the titanium plate wrapped with 25 μg/ml EMD treated-BMSC film into nude mice for 8 weeks, more nodules were formed on the surface of the titanium plate than that the control (P < 0.05). HE showed that there is a little blue-violet immature bone-like tissue block. Besides, the expression of RUNX Family Transcription Factor 2 (Runx2), Osterix, Osteocalcin (OCN), collagen I (COLI), alkaline phosphatase (ALP) and β-catenin were inhibited in xav939 group (P < 0.05); Inversely, all were activated in EMD group (P < 0.05). In conclusion, EMD promoted the proliferation and osteogenic differentiation of BMSCs. EMD's function on BMSCs might be associated with the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Lu Cheng
- Department of Prosthodontics, Guiyang Hospital of Stomatology, Guiyang, Gsuizhou Province, 550002, People's Republic of China
| | - Ying Li
- Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - Qian Xia
- Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - MaoHua Meng
- Department of Prosthodontics, School of Stomatology, Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - ZhaoYang Ye
- Clinical Research Center, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - ZhengLong Tang
- Department of Prosthodontics, School of Stomatology, Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China.,Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - HongChao Feng
- Department of Oral and Maxillofacial Surgery, Guiyang Hospital of Stomatology, Guiyang, Guizhou Province, 550002, People's Republic of China
| | - Xin Chen
- Department of Prosthodontics, School of Stomatology, Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - HeLin Chen
- Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - Xiao Zeng
- Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - Yi Luo
- Department of Prosthodontics, Guiyang Hospital of Stomatology, Guiyang, Gsuizhou Province, 550002, People's Republic of China
| | - Qiang Dong
- Department of Prosthodontics, School of Stomatology, Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China.,Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| |
Collapse
|
30
|
SPRY4 acts as an indicator of osteoarthritis severity and regulates chondrocyte hypertrophy and ECM protease expression. NPJ Regen Med 2021; 6:56. [PMID: 34535669 PMCID: PMC8448831 DOI: 10.1038/s41536-021-00165-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 08/11/2021] [Indexed: 11/09/2022] Open
Abstract
Osteoarthritis (OA) causes serious changes in the metabolic and signaling pathways of chondrocytes, including the mitogen-activated protein kinase (MAPK) pathway. However, the role of sprouty RTK signaling antagonist 4 (SPRY4), an inhibitor of MAPK, in the human cartilage tissues and chondrocytes remains to be understood. Here, using SPRY4 gene delivery into healthy and degenerated chondrocytes, we elucidated the role of SPRY4 in preventing chondrocyte hypertrophy. In addition to using the human cartilage tissues with the destabilization of the medial meniscus (DMM) model in Sprague-Dawley (SD) rats, the role of SPRY4 in cartilage tissues and chondrocytes was explored through their molecular and histological analyses. In order to determine the effects of SPRY4 on healthy human chondrocyte hypertrophy, small interfering RNA (siRNA) was used to knock down SPRY4. Lentiviral transduction of SPRY4 into degenerated human chondrocytes allowed us to investigate its ability to prevent hypertrophy. SPRY4 expression levels were higher in healthy human cartilage tissue and chondrocytes than in degenerated human cartilage tissues and hypertrophy-induced chondrocytes. The knockdown of SPRY4 in healthy chondrocytes caused an increase in hypertrophy, senescence, reactive oxygen species (ROS) production, and extracellular matrix (ECM) protease expression. However, all these factors decreased upon overexpression of SPRY4 in degenerated chondrocytes via regulation of the MAPK signaling pathway. We conclude that SPRY4 is a crucial indicator of osteoarthritis (OA) severity and could play an important role in preventing OA in the cartilage by inhibiting chondrocyte hypertrophy.
Collapse
|
31
|
Biological Mechanisms of Paeonoside in the Differentiation of Pre-Osteoblasts and the Formation of Mineralized Nodules. Int J Mol Sci 2021; 22:ijms22136899. [PMID: 34199016 PMCID: PMC8268717 DOI: 10.3390/ijms22136899] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 12/23/2022] Open
Abstract
Paeonia suffruticosa is a magnificent and long-lived woody plant that has traditionally been used to treat various diseases including inflammatory, neurological, cancer, and cardiovascular diseases. In the present study, we demonstrated the biological mechanisms of paeonoside (PASI) isolated from the dried roots of P. suffruticosa in pre-osteoblasts. Herein, we found that PASI has no cytotoxic effects on pre-osteoblasts. Migration assay showed that PASI promoted wound healing and transmigration in osteoblast differentiation. PASI increased early osteoblast differentiation and mineralized nodule formation. In addition, PASI enhanced the expression of Wnt3a and bone morphogenetic protein 2 (BMP2) and activated their downstream molecules, Smad1/5/8 and β-catenin, leading to increases in runt-related transcription factor 2 (RUNX2) expression during osteoblast differentiation. Furthermore, PASI-mediated osteoblast differentiation was attenuated by inhibiting the BMP2 and Wnt3a pathways, which was accompanied by reduction in the expression of RUNX2 in the nucleus. Taken together, our findings provide evidence that PASI enhances osteoblast differentiation and mineralized nodules by regulating RUNX2 expression through the BMP2 and Wnt3a pathways, suggesting a potential role for PASI targeting osteoblasts to treat bone diseases including osteoporosis and periodontitis.
Collapse
|
32
|
Park KR, Lee JY, Cho M, Hong JT, Yun HM. Paeonolide as a Novel Regulator of Core-Binding Factor Subunit Alpha-1 in Bone-Forming Cells. Int J Mol Sci 2021; 22:ijms22094924. [PMID: 34066458 PMCID: PMC8125120 DOI: 10.3390/ijms22094924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 02/07/2023] Open
Abstract
Paeonia suffruticosa has been extensively used as a traditional medicine with various beneficial effects; paeonolide (PALI) was isolated from its dried roots. This study aimed to investigate the novel effects and mechanisms of PALI in pre-osteoblasts. Here, cell viability was evaluated using an MTT assay. Early and late osteoblast differentiation was examined by analyzing the activity of alkaline phosphatase (ALP) and by staining it with Alizarin red S (ARS). Cell migration was assessed using wound healing and Boyden chamber assays. Western blot and immunofluorescence analyses were used to examine the intracellular signaling pathways and differentiation proteins. PALI (0.1, 1, 10, 30, and 100 μM) showed no cytotoxic or proliferative effects in pre-osteoblasts. In the absence of cytotoxicity, PALI (1, 10, and 30 μM) promoted wound healing and transmigration during osteoblast differentiation. ALP staining demonstrated that PALI (1, 10, and 30 μM) promoted early osteoblast differentiation in a dose-dependent manner, and ARS staining showed an enhanced mineralized nodule formation, a key indicator of late osteoblast differentiation. Additionally, low concentrations of PALI (1 and 10 μM) increased the bone morphogenetic protein (BMP)–Smad1/5/8 and Wnt–β-catenin pathways in osteoblast differentiation. Particularly, PALI (1 and 10 μM) increased the phosphorylation of ERK1/2 compared with BMP2 treatment, an FDA-approved drug for bone diseases. Furthermore, PALI-mediated early and late osteoblast differentiation was abolished in the presence of the ERK1/2 inhibitor U0126. PALI-induced RUNX2 (Cbfa1) expression and nuclear localization were also attenuated by blocking the ERK1/2 pathway during osteoblast differentiation. We suggest that PALI has biologically novel activities, such as enhanced osteoblast differentiation and bone mineralization mainly through the intracellular ERK1/2-RUNX2 signaling pathway, suggesting that PALI might have therapeutic action and aid the treatment and prevention of bone diseases, such as osteoporosis and periodontitis.
Collapse
Affiliation(s)
- Kyung-Ran Park
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Korea;
| | - Joon Yeop Lee
- National Institute for Korean Medicine Development, Gyeongsan 38540, Korea; (J.Y.L.); (M.C.)
| | - Myounglae Cho
- National Institute for Korean Medicine Development, Gyeongsan 38540, Korea; (J.Y.L.); (M.C.)
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk 28160, Korea;
| | - Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Korea;
- Correspondence: ; Tel.: +82-02-961-0691; Fax: +82-02-960-1457
| |
Collapse
|
33
|
Low-dose IL-34 has no effect on osteoclastogenesis but promotes osteogenesis of hBMSCs partly via activation of the PI3K/AKT and ERK signaling pathways. Stem Cell Res Ther 2021; 12:268. [PMID: 33947456 PMCID: PMC8097863 DOI: 10.1186/s13287-021-02263-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/02/2021] [Indexed: 12/29/2022] Open
Abstract
Background Inflammatory microenvironment is significant to the differentiation and function of mesenchymal stem cells (MSCs). It evidentially influences the osteoblastogenesis of MSCs. IL-34, a newly discovered cytokine, playing a key role in metabolism. However, the research on its functional role in the osteogenesis of MSCs was rarely reported. Here, we described the regulatory effects of low-dose IL-34 on both osteoblastogenesis and osteoclastogenesis. Methods We performed the osteogenic effects of hBMSCs by exogenous and overexpressed IL-34 in vitro, so were the osteoclastogenesis effects of mBMMs by extracellular IL-34. CCK-8 was used to assess the effect of IL-34 on the viability of hBMSCs and mBMMs. ALP, ARS, and TRAP staining was used to evaluate ALP activity, mineral deposition, and osteoclastogenesis, respectively. qRT-PCR and Western blotting analysis were performed to detect the expression of target genes and proteins. ELISA was used to evaluate the concentrations of IL-34. In vivo, a rat tibial osteotomy model and an OVX model were established. Radiographic analysis and histological evaluation were performed to confirm the therapeutic effects of IL-34 in fracture healing and osteoporosis. Statistical differences were evaluated by two-tailed Student’s t test, one-way ANOVA with Bonferroni’s post hoc test, and two-way ANOVA with Bonferroni multiple comparisons post hoc test in the comparison of 2 groups, more than 2 groups, and different time points of treated groups, respectively. Results Promoted osteoblastogenesis of hBMSCs was observed after treated by exogenous or overexpressed IL-34 in vitro, confirmed by increased mineral deposits and ALP activity. Furthermore, exogenous or overexpressed IL-34 enhanced the expression of p-AKT and p-ERK. The specific AKT and ERK signaling pathway inhibitors suppressed the enhancement of osteoblastogenesis induced by IL-34. In a rat tibial osteotomy model, imaging and histological analyses testified the local injection of exogenous IL-34 improved bone healing. However, the additional IL-34 has no influence on both osteoclastogenesis of mBMMs in vitro and osteoporosis of OVX model of rat in vivo. Conclusions Collectively, our study demonstrate that low-dose IL-34 regulates osteogenesis of hBMSCs partly via the PIK/AKT and ERK signaling pathway and enhances fracture healing, with neither promoting nor preventing osteoclastogenesis in vitro and osteoporosis in vivo.
Collapse
|
34
|
Park KR, Lee JY, Cho M, Yun HM. Ziyuglycoside I Upregulates RUNX2 through ERK1/2 in Promoting Osteoblast Differentiation and Bone Mineralization. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:883-900. [PMID: 33829967 DOI: 10.1142/s0192415x21500427] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sanguisorba officinalis L. (Rosaceae) is a perennial herbaceous plant and its roots have been used as an important traditional medicine for over 2000 years. Ziyuglycoside I (Ziyu), an active compound isolated from the roots of S. officinalis L., has shown biological effects such as anti-oxidant, antiviral, and antiwrinkle activities. This study aimed to elucidate the underlying mechanisms of action of Ziyu on cytotoxicity, migration, and differentiation of pre-osteoblasts. Herein, at concentrations ranging from 1 to 100 [Formula: see text]M, Ziyu was not cytotoxic against pre-osteoblasts. Alkaline phosphatase activity assay and staining, and migration assay showed that Ziyu increased cell migration and promoted early osteoblast differentiation, followed by the enhancement of mineralized nodule formation in a dose-dependent manner, as indicated by Alizarin Red S staining. In addition, Ziyu increased the protein levels of runt-related transcription factor 2 (RUNX2) during osteoblast differentiation, whereas it did not affect the phosphorylation of Smad1/5/8 and GSK3b and expression of [Formula: see text]-catenin. Ziyu also activated ERK1/2 and mitogen-activated protein kinase during osteoblast differentiation, and ERK1/2 inhibitor attenuated Ziyu-mediated RUNX2 expression and nuclear accumulation. Furthermore, Ziyu-mediated early and late osteoblast differentiation was significantly suppressed by the inhibition of ERK1/2, which was accompanied by attenuation in the mRNA levels of osteoblast-related genes including bone sialoprotein, osteopontin, and osteocalcin. Taken together, the findings of this study provide evidence that Ziyu promotes cell migration, osteoblast differentiation, and bone mineralization and suggest a potential role for Ziyu in the treatment of bone diseases.
Collapse
Affiliation(s)
- Kyung-Ran Park
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Joon Yeop Lee
- National Institute for Korean Medicine Development, Gyeongsan 38540, Republic of Korea
| | - MyoungLae Cho
- National Institute for Korean Medicine Development, Gyeongsan 38540, Republic of Korea
| | - Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02453, Republic of Korea
| |
Collapse
|
35
|
Zhu Y, Ortiz A, Costa M. Wrong place, wrong time: Runt-related transcription factor 2/SATB2 pathway in bone development and carcinogenesis. J Carcinog 2021; 20:2. [PMID: 34211338 PMCID: PMC8202446 DOI: 10.4103/jcar.jcar_22_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/03/2020] [Accepted: 01/06/2021] [Indexed: 12/23/2022] Open
Abstract
Upregulation or aberrant expression of genes such as special AT-rich sequence-binding protein 2 (SATB2) is necessary for normal cell differentiation and tissue development and is often associated with carcinogenesis and metastatic progression. SATB2 is a critical transcription factor for biological development of various specialized cell lineages, such as osteoblasts and neurons. The dysregulation of SATB2 expression has recently been associated with various types of cancer, while the mechanisms and pathways by which it mediates tumorigenesis are not well elucidated. Runt-related transcription factor 2 (RUNX2) is a master regulator for osteogenesis, and it shares common pathways with SATB2 to regulate bone development. Interestingly, these two transcription factors co-occur in several epithelial and mesenchymal cancers and are linked by multiple cancer-related proteins and microRNAs. This review examines the interactions between RUNX2 and SATB2 in a network necessary for normal bone development and the circumstances in which the expression of RUNX2 and SATB2 in the wrong place and time leads to carcinogenesis.
Collapse
Affiliation(s)
- Yusha Zhu
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| | - Angelica Ortiz
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| | - Max Costa
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
36
|
Crous A, Abrahamse H. The Signalling Effects of Photobiomodulation on Osteoblast Proliferation, Maturation and Differentiation: A Review. Stem Cell Rev Rep 2021; 17:1570-1589. [PMID: 33686595 DOI: 10.1007/s12015-021-10142-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2021] [Indexed: 02/06/2023]
Abstract
Proliferation of osteoblasts is essential for maturation and mineralization of bone matrix. Ossification, the natural phase of bone-forming and hardening is a carefully regulated phase where deregulation of this process may result in insufficient or excessive bone mineralization or ectopic calcification. Osteoblasts can also be differentiated into osteocytes, populating short interconnecting passages within the bone matrix. Over the past few decades, we have seen a significant improvement in awareness and techniques using photobiomodulation (PBM) to stimulate cell function. One of the applications of PBM is the promotion of osteoblast proliferation and maturation. PBM research results on osteoblasts showed increased mitochondrial ATP production, increased osteoblast activity and proliferation, increased and pro-osteoblast expression in the presence of red and NIR radiation. Osteocyte differentiation was also accomplished using blue and green light, showing that different light parameters have various signalling effects. The current review addresses osteoblast function and control, a new understanding of PBM on osteoblasts and its therapeutic impact using various parameters to optimize osteoblast function that may be clinically important. Graphical Abstract.
Collapse
Affiliation(s)
- Anine Crous
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, PO Box 17011, Johannesburg, 2028, South Africa.
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, PO Box 17011, Johannesburg, 2028, South Africa
| |
Collapse
|
37
|
Ahuja N, Awad KR, Brotto M, Aswath PB, Varanasi V. A comparative study on silicon nitride, titanium and polyether ether ketone on mouse pre-osteoblast cells. MEDICAL DEVICES & SENSORS 2021; 4:e10139. [PMID: 35765350 PMCID: PMC9236125 DOI: 10.1002/mds3.10139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The current study provides more insights about the surface bioactivity of the silicon nitride (Si3N4) as a potential candidate for bone regeneration in craniofacial and orthopaedic applications compared with conventional implantation materials. Current skeletal reconstructive materials such as titanium and polyether ether ketone (PEEK) are limited by poor long-term stability, biocompatibility and prolonged healing. Si3N4 is an FDA-approved material for an intervertebral spacer in spinal fusion applications. It is biocompatible and has antimicrobial properties. Here, we hypothesize that Si3N4 was found to be an osteoconductive material and conducts the growth, differentiation of MC3T3-E1 cells for extracellular matrix deposition, mineralization and eventual bone regeneration for craniofacial and orthopaedic applications. MC3T3-E1 cells were used to study the osteoblastic differentiation and mineralization on sterile samples of Si3N4, titanium alloy and PEEK. The samples were then analysed for extracellular matrix deposition and mineralization by FTIR, Raman spectroscopy, SEM, EDX, Alizarin Red, qRT-PCR and ELISA. The in vitro study indicates the formation of collagen fibres and mineral deposition on all three sample surfaces. There was more profound and faster ECM deposition and mineralization on Si3N4 surface as compared to titanium and PEEK. The FTIR and Raman spectroscopy show formation of collagen and mineral deposition at 30 days for Si3N4 and titanium and not PEEK. The peaks shown by Raman for Si3N4 resemble closely to natural bone. Results also indicate the upregulation of osteogenic transcription factors such as RUNX2, SP7, collagen type I and osteocalcin. The authors concluded that Si3N4 rapidly conducts mineralized tissue formation via extracellular matrix deposition and biomarker expression in mouse calvarial pre-osteoblast cells. Thus, this study confirms that the bioactive Si3N4 could be a potential material for craniofacial and orthopaedic applications leading to rapid bone regeneration that resemble the natural bone structure.
Collapse
Affiliation(s)
- Neelam Ahuja
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, USA
| | - Kamal R. Awad
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, USA
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, TX, USA
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, USA
| | - Pranesh B Aswath
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, TX, USA
| | - Venu Varanasi
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, USA
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
38
|
Sánchez-de-Diego C, Pedrazza L, Pimenta-Lopes C, Martinez-Martinez A, Dahdah N, Valer JA, Garcia-Roves P, Rosa JL, Ventura F. NRF2 function in osteocytes is required for bone homeostasis and drives osteocytic gene expression. Redox Biol 2020; 40:101845. [PMID: 33373776 PMCID: PMC7773566 DOI: 10.1016/j.redox.2020.101845] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022] Open
Abstract
Osteocytes, the most abundant bone cell type, are derived from osteoblasts through a process in which they are embedded in an osteoid. We previously showed that nutrient restriction promotes the osteocyte transcriptional program and is associated with increased mitochondrial biogenesis. Here, we show that increased mitochondrial biogenesis increase reactive oxygen species (ROS) levels and consequently, NRF2 activity during osteocytogenesis. NRF2 activity promotes osteocyte-specific expression of Dmp1, Mepe, and Sost in IDG-SW3 cells, primary osteocytes, and osteoblasts, and in murine models with Nfe2l2 deficiency in osteocytes or osteoblasts. Moreover, ablation of Nfe2l2 in osteocytes or osteoblasts generates osteopenia and increases osteoclast numbers with marked sexual dimorphism. Finally, treatment with dimethyl fumarate prevented the deleterious effects of ovariectomy in trabecular bone masses of mice and restored osteocytic gene expression. Altogether, we uncovered the role of NRF2 activity in osteocytes during the regulation of osteocyte gene expression and maintenance of bone homeostasis. ROS levels and NRF2 activity are increased during osteocytogenesis. NRF2 drives osteocyte specification and activate the transcription of osteocyte-specific genes. NRF2 in osteocytes has a fundamental role in bone homeostasis and its deletion induces osteopenia. Activation of NRF2 with dimethyl fumarate prevents osteopenia induced by ovariectomy.
Collapse
Affiliation(s)
- Cristina Sánchez-de-Diego
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Leonardo Pedrazza
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Carolina Pimenta-Lopes
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Arturo Martinez-Martinez
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Norma Dahdah
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - José Antonio Valer
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Pablo Garcia-Roves
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Jose Luis Rosa
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
39
|
Effects of PIN on Osteoblast Differentiation and Matrix Mineralization through Runt-Related Transcription Factor. Int J Mol Sci 2020; 21:ijms21249579. [PMID: 33339165 PMCID: PMC7765567 DOI: 10.3390/ijms21249579] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Styrax Japonica Sieb. et Zucc. has been used as traditional medicine in inflammatory diseases, and isolated compounds have shown pharmacological activities. Pinoresinol glucoside (PIN) belonging to lignins was isolated from the stem bark of S. Japonica. This study aimed to investigate the biological function and mechanisms of PIN on cell migration, osteoblast differentiation, and matrix mineralization. Herein, we investigated the effects of PIN in MC3T3-E1 pre-osteoblasts, which are widely used for studying osteoblast behavior in in vitro cell systems. At concentrations ranging from 0.1 to 100 μM, PIN had no cell toxicity in pre-osteoblasts. Pre-osteoblasts induced osteoblast differentiation, and the treatment of PIN (10 and 30 μM) promoted the cell migration rate in a dose-dependent manner. At concentrations of 10 and 30 μM, PIN elevated early osteoblast differentiation in a dose-dependent manner, as indicated by increases in alkaline phosphatase (ALP) staining and activity. Subsequently, PIN also increased the formation of mineralized nodules in a dose-dependent manner, as indicated by alizarin red S (ARS) staining, demonstrating positive effects of PIN on late osteoblast differentiation. In addition, PIN induced the mRNA level of BMP2, ALP, and osteocalcin (OCN). PIN also upregulated the protein level of BMP2 and increased canonical BMP2 signaling molecules, the phosphorylation of Smad1/5/8, and the protein level of Runt-related transcription factor 2 (RUNX2). Furthermore, PIN activated non-canonical BMP2 signaling molecules, activated MAP kinases, and increased β-catenin signaling. The findings of this study indicate that PIN has biological roles in osteoblast differentiation and matrix mineralization, and suggest that PIN might have anabolic effects in bone diseases such as osteoporosis and periodontitis.
Collapse
|
40
|
Babaki D, Yaghoubi S, Matin MM. The effects of mineral trioxide aggregate on osteo/odontogenic potential of mesenchymal stem cells: a comprehensive and systematic literature review. Biomater Investig Dent 2020; 7:175-185. [PMID: 33313519 PMCID: PMC7717865 DOI: 10.1080/26415275.2020.1848432] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/07/2020] [Indexed: 12/12/2022] Open
Abstract
The significance of dental materials in dentin-pulp complex tissue engineering is undeniable. The mechanical properties and bioactivity of mineral trioxide aggregate (MTA) make it a promising biomaterial for future stem cell-based endodontic therapies. There are numerous in vitro studies suggesting the low cytotoxicity of MTA towards various types of cells. Moreover, it has been shown that MTA can enhance mesenchymal stem cells' (MSCs) osteo/odontogenic ability. According to the preferred reporting items for systematic reviews and meta-analyses (PRISMA), a literature review was conducted in the Medline, PubMed, and Scopus databases. Among the identified records, the cytotoxicity and osteo/odontoblastic potential of MTA or its extract on stem cells were investigated. Previous studies have discovered the differentiation-inducing potential of MTA on MSCs, providing a background for dentin-pulp complex cell therapies using the MTA, however, animal trials are needed before moving into clinical trials. In conclusion, MTA can be a promising candidate dental biomaterial for futuristic stem cell-based endodontic therapies.
Collapse
Affiliation(s)
- Danial Babaki
- Department of Biomedical Engineering, Tagliatela College of Engineering, University of New Haven, West Haven, CT, USA
| | - Sanam Yaghoubi
- Visiting Scholar at Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Maryam M. Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
41
|
Kang MA, Lee J, Park SH. Cannabidiol induces osteoblast differentiation via angiopoietin1 and p38 MAPK. ENVIRONMENTAL TOXICOLOGY 2020; 35:1318-1325. [PMID: 32656944 DOI: 10.1002/tox.22996] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 06/11/2023]
Abstract
In this study, we report the potential of cannabidiol, one of the major cannabis constituents, for enhancing osteoblastic differentiation in U2OS and MG-63 cells. Cannabidiol increased the expression of Angiopoietin1 and the enzyme activity of alkaline phosphatase in U2OS and MG-63. Invasion and migration assay results indicated that the cell mobility was activated by cannabidiol in U2OS and MG-63. Western blotting analysis showed that the expression of tight junction related proteins such as Claudin1, Claudin4, Occuludin1, and ZO1 was increased by cannabidiol in U2OS and MG-63. Alizarin Red S staining analysis showed that calcium deposition and mineralization was enhanced by cannabidiol in U2OS and MG-63. Western blotting analysis indicated that the expression of osteoblast differentiation related proteins such as distal-less homeobox 5, bone sialoprotein, osteocalcin, type I collagen, Runt-related transcription factor 2 (RUNX2), osterix (OSX), and alkaline phosphatase was time dependently upregulated by cannabidiol in U2OS and MG-63. Mechanistically, cannabidiol-regulated osteoblastic differentiation in U2OS and MG-63 by strengthen the protein-protein interaction among RUNX2, OSX, or the phosphorylated p38 mitogen-activated protein kinase (MAPK). In conclusion, cannabidiol increased Angiopoietin1 expression and p38 MAPK activation for osteoblastic differentiation in U2OS and MG-63 suggesting that cannabidiol might provide a novel therapeutic option for the bone regeneration.
Collapse
Affiliation(s)
- Mi-Ae Kang
- Department of Biological Science, Gachon University, Seongnam, Republic of Korea
| | - Jongsung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, Republic of Korea
| |
Collapse
|
42
|
Liu J, Yu X, Liu B, Yu H, Li Z. Phosphorylated MAPK14 Promotes the Proliferation and Migration of Bladder Cancer Cells by Maintaining RUNX2 Protein Abundance. Cancer Manag Res 2020; 12:11371-11382. [PMID: 33204153 PMCID: PMC7661795 DOI: 10.2147/cmar.s274058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/21/2020] [Indexed: 01/20/2023] Open
Abstract
Background Mitogen-activated protein kinase 14 (MAPK14) acts as an integration point for multiple biochemical signal pathways. High expressions of MAPK14 have been found in a variety of tumors. Runt‑related transcription factor 2 (RUNX2) is related to many tumors, especially in tumor invasion and metastasis. However, the mechanism of these two genes in bladder cancer remains unclear. Methods TCGA database and Western blot were used to analyze the mRNA and protein levels of the target gene in bladder cancer tissues and adjacent tissues. The proliferation ability of bladder cancer cells was tested by colony forming and EdU assay. The migration ability of cells was detected by transwell assay. Immunoprecipitation was utilized to detect protein-protein interaction. Cycloheximide chase assay was used to measure the half-life of RUNX2 protein. Results Phosphorylated mitogen-activated protein kinase 14 (P-MAPK14, Thr180/Tyr182) was highly expressed in bladder cancer tissues and bladder cancer cell lines. Accordingly, P-MAPK14 could be combined with RUNX2 and maintain its protein stability and promote the proliferation and migration of bladder cancer cells. In addition, the functional degradation caused by the downregulation of MAPK14 and P-MAPK14 could be partially compensated by the overexpression of RUNX2. Conclusion These results suggest that P-MAPK14 might play an important role in the development of bladder cancer and in the regulation of RUNX2 protein expression. P-MAPK14 might become a potential target for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Junlong Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| | - Xiuyue Yu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| | - Bitian Liu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, People's Republic of China
| | - Hongyuan Yu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| | - Zhenhua Li
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, People's Republic of China
| |
Collapse
|
43
|
Zhao LD, Xu WC, Cui J, Liang YC, Cheng WQ, Xin BC, Song J. Long non-coding RNA maternally expressed gene 3 inhibits osteogenic differentiation of human dental pulp stem cells via microRNA-543/smad ubiquitin regulatory factor 1/runt-related transcription factor 2 axis. Arch Oral Biol 2020; 118:104838. [PMID: 32711339 DOI: 10.1016/j.archoralbio.2020.104838] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The aim of the present study was to investigate the biological roles and underlying mechanism of the long non-coding RNA maternally expressed gene 3 (MEG3) on osteogenic differentiation of human dental pulp stem cells (hDPSCs). METHODS The expression levels of MEG3, microRNA-543 (miR-543), osterix, osteopontin, osteocalcin and runt-related transcription factor 2 (RUNX2) were measured by quantitative real-time PCR (qRT-PCR). Alkaline phosphatase (ALP) activity assay and alizarin red S staining (ARS) were used to measure the impacts exerted by MEG3, miR-543 on osteogenic differentiation. Cell proliferation was measured by MTT assay. In addition, the targeted relationships between miR-543, MEG3, and Smad ubiquitin regulatory factor 1 (SMURF1) were assessed through dual luciferase reporter assay. RESULTS During osteogenic induction, the expression of MEG3 was gradually reduced, whereas the expression of miR-543, osterix, osteopontin, osteocalcin and RUNX2 were gradually increased. Functional analysis implied that MEG3 overexpression or miR-543 inhibition reduced the cell proliferation, ALP activity, ARS levels, and decreased the expression of osteoblast-related proteins. Moreover, MEG3 promoted SMURF1 expression by directly targeting miR-543 as a competing endogenous RNA. Furthermore, overexpression of miR-543 or silencing SMURF1 could reverse the inhibitory effects of MEG3 on the osteogenic differentiation of hDPSCs. CONCLUSIONS In conclusion, our study revealed that overexpression of MEG3 inhibited hDPSCs osteogenic differentiation via miR-543/SMURF1/RUNX2 regulatory network, which may contribute to the functional regulation and clinical applications of hDPSCs.
Collapse
Affiliation(s)
- Luo-Dan Zhao
- Department of Oral and Maxillofacial Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangzhou Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Wei-Cheng Xu
- Department of Dental Implantology, Yantai Stomatological Hospital, Yantai, 264001, PR China
| | - Jian Cui
- Department of Dental Implantology, Yantai Stomatological Hospital, Yantai, 264001, PR China
| | - Yan-Can Liang
- Department of Oral and Maxillofacial Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangzhou Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Wei-Qi Cheng
- Department of Oral and Maxillofacial Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangzhou Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Bing-Chang Xin
- Department of Cariology and Endodontology, Qingdao Stomatological Hospital, Qingdao, 266001, PR China.
| | - Jia Song
- Department of Cariology and Endodontology, Qingdao Stomatological Hospital, Qingdao, 266001, PR China.
| |
Collapse
|
44
|
Hoshikawa S, Shimizu K, Watahiki A, Chiba M, Saito K, Wei W, Fukumoto S, Inuzuka H. Phosphorylation-dependent osterix degradation negatively regulates osteoblast differentiation. FASEB J 2020; 34:14930-14945. [PMID: 32931083 DOI: 10.1096/fj.202001340r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/15/2020] [Accepted: 08/25/2020] [Indexed: 01/05/2023]
Abstract
Proteasome inhibitors exert an anabolic effect on bone formation with elevated levels of osteoblast markers. These findings suggest the important role of the proteasomal degradation of osteogenic regulators, while the underlying molecular mechanisms are not fully understood. Here, we report that the proteasome inhibitors bortezomib and ixazomib markedly increased protein levels of the osteoblastic key transcription factor osterix/Sp7 (Osx). Furthermore, we revealed that Osx was targeted by p38 and Fbw7 for proteasomal degradation. Mechanistically, p38-mediated Osx phosphorylation at S73/77 facilitated Fbw7 interaction to trigger subsequent Osx ubiquitination. Consistent with these findings, p38 knockdown or pharmacological p38 inhibition resulted in Osx protein stabilization. Treatment with p38 inhibitors following osteogenic stimulation efficiently induced osteoblast differentiation through Osx stabilization. Conversely, pretreatment of p38 inhibitor followed by osteogenic challenge impaired osteoblastogenesis via suppressing Osx expression, suggesting that p38 exerts dual but opposite effects in the regulation of Osx level to fine-tune its activity during osteoblast differentiation. Furthermore, Fbw7-depleted human mesenchymal stem cells and primary mouse calvarial cells resulted in increased osteogenic capacity. Together, our findings unveil the molecular mechanisms underlying the Osx protein stability control and suggest that targeting the Osx degradation pathway could help enhance efficient osteogenesis and bone matrix regeneration.
Collapse
Affiliation(s)
- Seira Hoshikawa
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan.,Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Kouhei Shimizu
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Asami Watahiki
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Mitsuki Chiba
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan.,Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Kan Saito
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Satoshi Fukumoto
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan.,Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Japan.,Department of Pediatric Dentistry, Kyushu University Graduate School of Dental Science, Fukuoka, Japan
| | - Hiroyuki Inuzuka
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
45
|
Kidwai F, Mui BWH, Arora D, Iqbal K, Hockaday M, de Castro Diaz LF, Cherman N, Martin D, Myneni VD, Ahmad M, Futrega K, Ali S, Merling RK, Kaufman DS, Lee J, Robey PG. Lineage-specific differentiation of osteogenic progenitors from pluripotent stem cells reveals the FGF1-RUNX2 association in neural crest-derived osteoprogenitors. Stem Cells 2020; 38:1107-1123. [PMID: 32442326 PMCID: PMC7484058 DOI: 10.1002/stem.3206] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/01/2020] [Indexed: 12/16/2022]
Abstract
Human pluripotent stem cells (hPSCs) can provide a platform to model bone organogenesis and disease. To reflect the developmental process of the human skeleton, hPSC differentiation methods should include osteogenic progenitors (OPs) arising from three distinct embryonic lineages: the paraxial mesoderm, lateral plate mesoderm, and neural crest. Although OP differentiation protocols have been developed, the lineage from which they are derived, as well as characterization of their genetic and molecular differences, has not been well reported. Therefore, to generate lineage-specific OPs from human embryonic stem cells and human induced pluripotent stem cells, we employed stepwise differentiation of paraxial mesoderm-like cells, lateral plate mesoderm-like cells, and neural crest-like cells toward their respective OP subpopulation. Successful differentiation, confirmed through gene expression and in vivo assays, permitted the identification of transcriptomic signatures of all three cell populations. We also report, for the first time, high FGF1 levels in neural crest-derived OPs-a notable finding given the critical role of fibroblast growth factors (FGFs) in osteogenesis and mineral homeostasis. Our results indicate that FGF1 influences RUNX2 levels, with concomitant changes in ERK1/2 signaling. Overall, our study further validates hPSCs' power to model bone development and disease and reveals new, potentially important pathways influencing these processes.
Collapse
Affiliation(s)
- Fahad Kidwai
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Byron W. H. Mui
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Deepika Arora
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
- Biosystems and Biomaterials DivisionNational Institute of Standards and TechnologyGaithersburgMarylandUSA
| | - Kulsum Iqbal
- Department of Health and Human ServicesDental Consult Services, National Institute of Health Dental ClinicBethesdaMarylandUSA
| | - Madison Hockaday
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Luis Fernandez de Castro Diaz
- Department of Health and Human ServicesSkeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Natasha Cherman
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Daniel Martin
- Department of Health and Human ServicesGenomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Vamsee D. Myneni
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch/Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Moaz Ahmad
- Department of Health and Human ServicesMolecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Katarzyna Futrega
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Sania Ali
- Biology of Global Health, Department of BiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
| | - Randall K. Merling
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Dan S. Kaufman
- Department of MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Janice Lee
- Department of Health and Human ServicesCraniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Pamela G. Robey
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
46
|
Liu X, Fan Y, Xie J, Zhang L, Li L, Wang Z. Dehydroandrographolide Inhibits Osteosarcoma Cell Growth and Metastasis by Targeting SATB2-mediated EMT. Anticancer Agents Med Chem 2020; 19:1728-1736. [PMID: 31284872 DOI: 10.2174/1871520619666190705121614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND The 12-hydroxy-14-dehydroandrographolide (DP) is a predominant component of the traditional herbal medicine Andrographis paniculata (Burm. f.) Nees (Acanthaceae). Recent studies have shown that DP exhibits potent anti-cancer effects against oral and colon cancer cells. OBJECTIVE This investigation examined the potential effects of DP against osteosarcoma cell. METHODS A cell analyzer was used to measure cell viability. The cell growth and proliferation were performed by Flow cytometry and BrdU incorporation assay. The cell migration and invasion were determined by wound healing and transwell assay. The expression of EMT related proteins was examined by Western blot analysis. RESULTS In this study, we found that DP treatment repressed osteosarcoma (OS) cell growth in a dose-dependent manner. DP treatment significantly inhibited OS cell proliferation by arresting the cell cycle at G2/M phase. In addition, DP treatment effectively inhibited the migration and invasion abilities of OS cells through wound healing and Transwell tests. Mechanistic studies revealed that DP treatment effectively rescued the epithelialmesenchymal transition (EMT), while forced expression of SATB2 in OS cells markedly reversed the pharmacological effect of DP on EMT. CONCLUSION Our data demonstrated that DP repressed OS cell growth through inhibition of proliferation and cell cycle arrest; DP also inhibited metastatic capability of OS cells through a reversal of EMT by targeting SATB2. These findings demonstrate DP's potential as a therapeutic drug for OS treatment.
Collapse
Affiliation(s)
- Xuefeng Liu
- Department of Forensic Pathology, Xi'an Jiaotong University, School of Medicine, Xi'an, 710061, China.,Department of Anatomy, Jinzhou Medical University, Jinzhou, 121001, China
| | - Yonggang Fan
- Department of Cell Biology, Taizhou University, Taizhou, 375000, China
| | - Jing Xie
- Department of Cell Biology, Taizhou University, Taizhou, 375000, China
| | - Li Zhang
- Jinzhoushi Oral Cavity Hospital, Jinzhou, 121001, China
| | - Lihua Li
- Department of Cell Biology, Taizhou University, Taizhou, 375000, China
| | - Zhenyuan Wang
- Department of Forensic Pathology, Xi'an Jiaotong University, School of Medicine, Xi'an, 710061, China
| |
Collapse
|
47
|
Hojo H, Ohba S. Gene regulatory landscape in osteoblast differentiation. Bone 2020; 137:115458. [PMID: 32474244 DOI: 10.1016/j.bone.2020.115458] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/25/2020] [Accepted: 05/25/2020] [Indexed: 12/29/2022]
Abstract
The development of osteoblasts, a bone-forming cell population, occurs in conjunction with development of the skeleton, which creates our physical framework and shapes the body. In the past two decades, genetic studies have uncovered the molecular framework of this process-namely, transcriptional regulators and signaling pathways coordinate the cell fate determination and differentiation of osteoblasts in a spatial and temporal manner. Recently emerging genome-wide studies provide additional layers of understanding of the gene regulatory landscape during osteoblast differentiation, allowing us to gain novel insight into the modes of action of the key regulators, functional interaction among the regulator-bound enhancers, epigenetic regulations, and the complex nature of regulatory inputs. In this review, we summarize current understanding of the transcriptional regulation in osteoblasts, in terms of the gene regulatory landscape.
Collapse
Affiliation(s)
- Hironori Hojo
- Department of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Shinsuke Ohba
- Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan.
| |
Collapse
|
48
|
Zhang Y, Zhou K, Wu L, Gu H, Huang Z, Xu J. Downregulation of microRNA‑143 promotes osteogenic differentiation of human adipose‑derived mesenchymal stem cells through the k‑Ras/MEK/ERK signaling pathway. Int J Mol Med 2020; 46:965-976. [PMID: 32582994 PMCID: PMC7388841 DOI: 10.3892/ijmm.2020.4651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 11/22/2019] [Indexed: 12/29/2022] Open
Abstract
MicroRNAs (miRNAs) are known to have regulatory roles in the osteogenic differentiation of various mesenchymal stem cells (MSCs), although their regulatory role on human adipose‑derived mesenchymal stem cells (hADSCs) remains unclear. The aim of the present study was to investigate the biological function and underlying molecular mechanism of miRNAs in regulating the osteogenic differentiation of hADSCs using microarray assay. hADSCs differentiated into osteoblasts under culture with osteogenic medium, with an increase observed in calcium deposits and alkaline phosphatase activity. The mRNA levels of bone sialoprotein, osteopontin and osteocalcin increased, whereas Runt‑related transcription factor‑2 expression decreased during osteogenic differentiation. In addition, miR‑143 was markedly downregulated during osteogenic differentiation, while miR‑143 overexpression inhibited and miR‑143 knockdown enhanced this process. miR‑143 overexpression also blocked extracellular signal‑regulated kinase 1/2 (ERK1/2) pathway activation, while miR‑143 inhibition enhanced it. The promoting effects of miR‑143 knockdown on the osteogenic differentiation of hADSCs were partly diminished by the mitogen‑activated protein kinase (MEK) inhibitors U0126 and PD98059. Bioinformatics analysis further revealed that miR‑143 targets k‑Ras and directly binds to the 3'‑untranslated region of its mRNA. Inhibition of miR‑143 enhanced the activation of the k‑Ras/MEK/ERK pathway during osteogenic differentiation, whereas miR‑143 overexpression had the opposite effect. Collectively, these results demonstrated that miR‑143 negatively regulates the osteogenic differentiation of hADSCs through the k‑Ras/MEK/ERK pathway, providing further insight into the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Kaifeng Zhou
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Liang Wu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Huijie Gu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Zhongyue Huang
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Jun Xu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| |
Collapse
|
49
|
Wang Z, Liu Q, Liu C, Tan W, Tang M, Zhou X, Sun T, Deng Y. Mg 2+ in β-TCP/Mg-Zn composite enhances the differentiation of human bone marrow stromal cells into osteoblasts through MAPK-regulated Runx2/Osx. J Cell Physiol 2020; 235:5182-5191. [PMID: 31742679 DOI: 10.1002/jcp.29395] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/27/2019] [Indexed: 12/15/2022]
Abstract
Inducing the osteogenic differentiation from bone marrow stromal cells (BMSCs) might be a potent strategy for treating bone loss and nonunion during fracture and improving fracture healing. Among several signaling pathways involved, mitogen-activated protein kinases (MAPKs) have been reported to play a critical role. Magnesium (Mg)-based alloys, including Mg-Zn alloy, have been used clinically as implants in the musculoskeletal field and could promote BMSC osteogenic differentiation. However, the underlying mechanisms remain unclear. In this study, we produced Mg-Zn alloy consists of Mg and low concentrations of Zn, calcium carbonate, and β-tricalcium phosphate (β-TCP; manifesting process not shown), prepared Mg, Zn, and Mg-Zn extracts, and investigated the specific effects of these extracts on human BMSC (hBMSC) osteogenic differentiation and MAPK signaling. Mg extracts and Mg-Zn extracts could significantly promote the osteogenic differentiation of hBMSCs as manifested as increased alkaline phosphatase levels, enhanced calcium nodules formation, and increased messenger RNA expression and protein levels of osteogenesis markers, including BMPs, Col-I, Runx2, and Osx; in the meantime, Mg culture medium (CM) and Mg-Zn CM both significantly enhanced the activation of MAPK signaling in hBMSCs. By adding ERK1/2 signaling, p38 signaling, or JNK signaling inhibitor to Mg-Zn CM, or conducting p38 MAPK silence in hBMSCs, we revealed that these extracts might promote hBMSC osteogenic differentiation via p38 MAPK signaling and MAPK-regulated Runx2/Osx. In conclusion, Mg2+ in β-TCP/Mg-Zn extract promotes the osteogenic differentiation of hBMSCs via MAPK-regulated Runx2/Osx interaction.
Collapse
Affiliation(s)
- Zhenting Wang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Urology Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Qing Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Congcong Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Wei Tan
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Mingying Tang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaohua Zhou
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tianshi Sun
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Youwen Deng
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| |
Collapse
|
50
|
Zhang T, Gao Y, Cui W, Li Y, Xiao D, Zhou R. Nanomaterials-based Cell Osteogenic Differentiation and Bone Regeneration. Curr Stem Cell Res Ther 2020; 16:36-47. [PMID: 32436831 DOI: 10.2174/1574888x15666200521083834] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/07/2020] [Accepted: 01/16/2020] [Indexed: 02/08/2023]
Abstract
With the rapid development of nanotechnology, various nanomaterials have been applied to bone repair and regeneration. Due to the unique chemical, physical and mechanical properties, nanomaterials could promote stem cells osteogenic differentiation, which has great potentials in bone tissue engineering and exploiting nanomaterials-based bone regeneration strategies. In this review, we summarized current nanomaterials with osteo-induction ability, which could be potentially applied to bone tissue engineering. Meanwhile, the unique properties of these nanomaterials and their effects on stem cell osteogenic differentiation are also discussed. Furthermore, possible signaling pathways involved in the nanomaterials- induced cell osteogenic differentiation are also highlighted in this review.
Collapse
Affiliation(s)
- Tianxu Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yang Gao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Weitong Cui
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yanjing Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ronghui Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|