1
|
Sun L, Dong R, Zhang S, Zhu J, Zheng L, Zhang J. Zoledronic Acid-Treated Rats Show Altered Jaw Histology and Gene Expression in Nonexposed Medication-Related Osteonecrosis of the Jaws. J Oral Maxillofac Surg 2024:S0278-2391(24)00797-3. [PMID: 39393783 DOI: 10.1016/j.joms.2024.08.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND Diagnosis and management of medication-related osteonecrosis of the jaws (MRONJ) prior to clinical exposure induced by trauma may lead to improved patient management. Currently, few studies have examined early histologic and molecular MRONJ-related changes in the jaws. PURPOSE This study aimed to identify histological and gene expression changes in the maxilla and mandible of Sprague-Dawley (SD) rats after zoledronic acid (ZA) treatment. STUDY DESIGN, SETTING, AND SAMPLE This was an in vivo animal study. The experiments were conducted in the laboratory at the Stomatology Hospital of Tianjin Medical University. A total of 12 SD rats were included. PREDICTOR VARIABLE The predictor variable was ZA exposure. Twelve SD rats were divided into 2 groups: experimental (n = 6) and control (n = 6), and they were intraperitoneally injected with ZA and saline, respectively. MAIN OUTCOME VARIABLE The outcome variables were histological and molecular changes. The maxilla, mandible, and ilium bone tissue samples were examined using Masson's trichrome and hematoxylin-eosin staining. Gene expression changes were identified using transcriptome sequencing, the Kyoto encyclopedia of genes and genomes, and gene interactome network analysis. The key changes were validated using the quantitative real-time polymerase chain reaction and immunohistochemistry. COVARIATES None. ANALYSES The t-test, χ2 test, and Fisher's exact probability method were used for statistical analyses using the Statistical Package for the Social Sciences software (version 26.0). RESULTS All animals remained healthy during the experiments. Histological staining revealed that the percentage of empty bone lacunae in the maxilla and mandible was significantly higher than that in the ilium (P < .01). In total, 552 genes were screened using transcriptome sequencing. The sonic hedgehog (Shh) signaling pathway was highly enriched. The key gene for the Shh interaction was distal-less homeobox 5. The Shh, distal-less homeobox 5, and bone morphogenetic protein 2 genes and protein expression levels in the maxilla and mandible were higher in the experimental group than in the control group (P < .05). CONCLUSION AND RELEVANCE MRONJ-induced osteonecrosis and gene expression changes precede trauma-induced clinical changes in the SD rat model. These findings may provide additional support for timely and clinically early diagnosis and intervention.
Collapse
Affiliation(s)
- Lijun Sun
- Department of Stomatology, The First Affiliated Hospital of Shihezi University, Xinjiang, China
| | - Rui Dong
- Department of Oral and Maxillofacial Surgery, Stomatology Hospital of Tianjin Medical University, Tianjin, China
| | - Shihan Zhang
- Department of Oral and Maxillofacial Surgery, Stomatology Hospital of Hebei Medical University, Hebei, China
| | - Jiaqi Zhu
- Department of Oral and Maxillofacial Surgery,Yinchuan Stomatology Hospital, Ningxia, China
| | - Lingjie Zheng
- Department of Stomatology, Fuyang People's Hospital, Anhui, China
| | - Jian Zhang
- Department of Oral and Maxillofacial Surgery, Stomatology Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
2
|
Seki Y, Takebe H, Nakao Y, Sato K, Mizoguchi T, Nakamura H, Iijima M, Hosoya A. Osteoblast differentiation of Gli1⁺ cells via Wnt and BMP signaling pathways during orthodontic tooth movement. J Oral Biosci 2024; 66:373-380. [PMID: 38499228 DOI: 10.1016/j.job.2024.03.004] [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: 01/24/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/20/2024]
Abstract
OBJECTIVES Factors that induce bone formation during orthodontic tooth movement (OTM) remain unclear. Gli1 was recently identified as a stem cell marker in the periodontal ligament (PDL). Therefore, we evaluated the mechanism of differentiation of Cre/LoxP-mediated Gli1/Tomato+ cells into osteoblasts during OTM. METHODS After the final administration of tamoxifen to 8-week-old Gli1-CreERT2/ROSA26-loxP-stop-loxP-tdTomato mice for 2 days, nickel-titanium closed coil springs were attached between the upper anterior alveolar bone and the first molar. Immunohistochemical localizations of β-catenin, Smad4, and Runx2 were observed in the PDL on 2, 5, and 10 days after OTM initiation. RESULTS In the untreated tooth, few Gli1/Tomato+ cells were detected in the PDL. Two days after OTM initiation, the number of Gli1/Tomato+ cells increased in the PDL on the tension side. On this side, 49.3 ± 7.0% of β-catenin+ and 48.7 ± 5.7% of Smad4+ cells were found in the PDL, and Runx2 expression was detected in some Gli1/Tomato+ cells apart from the alveolar bone. The number of positive cells in the PDL reached a maximum on day 5. In contrast, on the compression side, β-catenin and Smad4 exhibited less immunoreactivity. On day 10, Gli1/Tomato+ cells were aligned on the alveolar bone on the tension side, with some expressing Runx2. CONCLUSIONS Gli1+ cells in the PDL differentiated into osteoblasts during OTM. Wnt and bone morphogenetic proteins signaling pathways may be involved in this differentiation.
Collapse
Affiliation(s)
- Yuri Seki
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Science University of Hokkaido. Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| | - Hiroaki Takebe
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Science University of Hokkaido. Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| | - Yuya Nakao
- Division of Orthodontics and Dentofacial Orthopedics, Department of Oral Growth and Development, School of Dentistry, Health Science University of Hokkaido. Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| | - Kohei Sato
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| | | | - Hiroaki Nakamura
- Department of Oral Anatomy, Matsumoto Dental University, Nagano, 399-0781, Japan
| | - Masahiro Iijima
- Division of Orthodontics and Dentofacial Orthopedics, Department of Oral Growth and Development, School of Dentistry, Health Science University of Hokkaido. Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| | - Akihiro Hosoya
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Science University of Hokkaido. Ishikari-Tobetsu, Hokkaido, 061-0293, Japan.
| |
Collapse
|
3
|
Wang Y, Xiao M, Cai F, Li Y, Shi T, Zhou X, Tian S, Huang D. Roxadustat ameliorates vascular calcification in CKD rats by regulating HIF-2α/HIF-1α. ENVIRONMENTAL TOXICOLOGY 2024; 39:2363-2373. [PMID: 38156404 DOI: 10.1002/tox.24116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/08/2023] [Accepted: 12/10/2023] [Indexed: 12/30/2023]
Abstract
Vascular calcification (VC) is a common complication of chronic kidney disease (CKD). VC is a gene-regulated process similar to osteogenic differentiation. There are still no convincing schemes to prevent and reduce the development of VC. It has been reported that hypoxia-inducing factor 1α (HIF-1α) and endothelin-1(ET-1) are related to VC. In this study, we found that the expression of ET-1 and HIF-1α was enhanced after VC, the interaction between HIF-1α and ET-1 was confirmed by CO-IP and luciferase experiments. We found that ET-1 was an upregulated differential gene of calcified vascular smooth muscle cells (VSMCs) through gene sequencing. However, hypoxia-inducing factor 2α (HIF-2α) and HIF-1α have antagonistic effects on each other. HIF-1α is a pro-inflammatory cytokine, and HIF-2α can improve inflammation and fibrosis. Roxadustat, as a selective PHD3 inhibitor, preferentially activates HIF-2α. It is still unclear whether roxadustat improves VC in CKD by regulating the expression of HIF-2α/HIF-1α. Alizarin red staining and western blot as well as immunohistochemical results showed that roxadustat could significantly reduce the degree of vascular and VSMCs calcification in CKD rats. Serum HIF-1α and ET-1 were significantly decreased after roxadustat treatment. In addition, western blot results showed that roxadustat could decrease the expression of HIF-1α and ET-1 in vascular tissues and calcified VSMC, but HIF-2α expression significantly increased. Interestingly, our study confirmed that activation of HIF-1α or inhibition of HIF-2α reversed the ameliorating effect of roxadustat on VC, proving that the effect mediated by roxadustat is HIF-2α/HIF-1α dependent. We have demonstrated for the first time that roxadustat improves VC in CKD rats by regulating HIF-2α/HIF-1α, thus providing a new idea for the application of roxadustat in VC of CKD.
Collapse
Affiliation(s)
- Yujing Wang
- Department of Hemodialysis, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Min Xiao
- Department of Clinical Laboratory, Strategic Support Force Medical Center, Beijing, China
| | - Feng Cai
- Department of Scientific Research, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Yang Li
- Department of Nephrology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Tianli Shi
- Department of Nephrology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Xiaoyan Zhou
- Department of Hemodialysis, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Shuhong Tian
- Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou, China
| | - Denggao Huang
- Department of Central Laboratory, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| |
Collapse
|
4
|
Dashti P, Lewallen EA, Gordon JAR, Montecino MA, Davie JR, Stein GS, van Leeuwen JPTM, van der Eerden BCJ, van Wijnen AJ. Epigenetic regulators controlling osteogenic lineage commitment and bone formation. Bone 2024; 181:117043. [PMID: 38341164 DOI: 10.1016/j.bone.2024.117043] [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: 11/07/2023] [Revised: 01/08/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
Bone formation and homeostasis are controlled by environmental factors and endocrine regulatory cues that initiate intracellular signaling pathways capable of modulating gene expression in the nucleus. Bone-related gene expression is controlled by nucleosome-based chromatin architecture that limits the accessibility of lineage-specific gene regulatory DNA sequences and sequence-specific transcription factors. From a developmental perspective, bone-specific gene expression must be suppressed during the early stages of embryogenesis to prevent the premature mineralization of skeletal elements during fetal growth in utero. Hence, bone formation is initially inhibited by gene suppressive epigenetic regulators, while other epigenetic regulators actively support osteoblast differentiation. Prominent epigenetic regulators that stimulate or attenuate osteogenesis include lysine methyl transferases (e.g., EZH2, SMYD2, SUV420H2), lysine deacetylases (e.g., HDAC1, HDAC3, HDAC4, HDAC7, SIRT1, SIRT3), arginine methyl transferases (e.g., PRMT1, PRMT4/CARM1, PRMT5), dioxygenases (e.g., TET2), bromodomain proteins (e.g., BRD2, BRD4) and chromodomain proteins (e.g., CBX1, CBX2, CBX5). This narrative review provides a broad overview of the covalent modifications of DNA and histone proteins that involve hundreds of enzymes that add, read, or delete these epigenetic modifications that are relevant for self-renewal and differentiation of mesenchymal stem cells, skeletal stem cells and osteoblasts during osteogenesis.
Collapse
Affiliation(s)
- Parisa Dashti
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Eric A Lewallen
- Department of Biological Sciences, Hampton University, Hampton, VA, USA
| | | | - Martin A Montecino
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad Andres Bello, Santiago, Chile; Millennium Institute Center for Genome Regulation (CRG), Santiago, Chile
| | - James R Davie
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada; CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, Manitoba R3E 0V9, Canada.
| | - Gary S Stein
- Department of Biochemistry, University of Vermont, Burlington, VT, USA
| | | | - Bram C J van der Eerden
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands.
| | - Andre J van Wijnen
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands; Department of Biochemistry, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
5
|
Wang Y, Han W, Zhong Y, Li W, Liu Q. Calcitriol combined with high-calcium and high-phosphorus diet induces vascular calcification model in chronic kidney disease rats. ENVIRONMENTAL TOXICOLOGY 2024; 39:1769-1779. [PMID: 38064270 DOI: 10.1002/tox.24039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/18/2023] [Accepted: 10/31/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND Cardiovascular diseases represent a significant complication arising from chronic kidney disease (CKD). Vascular calcification is an important risk factor for cardiovascular diseases. Reducing vascular calcification is therefore critical to reducing mortality in CKD patients. HYPOTHESIS This study aims to establish a vascular calcification model in rats with CKD by administering subcutaneous injections of calcitriol in combination with a high-calcium and high-phosphorus diet. METHODS The rats were divided into the CKD vascular calcification model group (subtotal nephrectomy+ [SNx+]) and the sham-operated control group (subtotal nephrectomy- [SNx-]). The rats in the SNx(+) group were administered high-calcium and high-phosphorus feeds following a 5/6 nephrectomy. Calcitriol (1 μg/kg, three times a week) was injected subcutaneously at weeks 0, 4, 8, and 12 after the operation. Measurements of body weight, urine, serum biochemical indicators and vascular calcification level were conducted in rats. RESULTS (1) Compared with the SNx(-) group, rats in the SNx(+) group experienced an increase in 24-h urine output, urinary phosphorus, and urinary microprotein excretion, along with the development of severe anemia. Additionally, there was a notable elevation in serum phosphorus, blood urea nitrogen, blood creatinine, fibroblast growth factor 23 (FGF-23), and intact parathyroid hormone levels, accompanied by severe hypoproteinemia at week 12. (2) The results of micro-compuyed tomography (μCT) and alizarin S staining of the thoracic aorta demonstrated an increase in vascular calcification in the SNx(+) group. (3) The expression levels of vascular calcification-related proteins were increased. CONCLUSIONS The administration of calcitriol combined with a high-calcium and high-phosphorus diet was found to induce vascular calcification in CKD rats, leading to a disturbance in mineral metabolism. Vascular calcification was effectively induced in CKD rats after 12 weeks of modeling, thereby presenting a novel approach for establishing a vascular calcification model in CKD rats, helping to elucidate this clinical condition and its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Yujing Wang
- Department of Hemodialysis, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Wenlong Han
- Department of Pharmacology, Hainan Medical University, Haikou, China
| | - Yuxiang Zhong
- Department of Hemodialysis, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Wenning Li
- Department of Nephrology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qiang Liu
- Department of Pharmacology, Hainan Medical University, Haikou, China
| |
Collapse
|
6
|
Chen CF, Wang PF, Liao HT. Platelet-Rich Plasma Lysate Enhances the Osteogenic Differentiation of Adipose-Derived Stem Cells. Ann Plast Surg 2024; 92:S12-S20. [PMID: 38285990 DOI: 10.1097/sap.0000000000003765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
ABSTRACT Adipose-derived stem cells (ADSCs) have become an accepted source of cells in bone tissue engineering. This study aimed to investigate whether platelet-rich plasma (PRP) lysate can replace traditional fetal bovine serum as a culture medium with the enhanced proliferation and osteogenic potential of ADSCs. We divided the experiment into 5 groups where the ADSCs were cultured in an osteogenic medium containing 2.5%, 5%, 7.5%, and 10% PRP lysate with 10% fetal bovine serum as the control group. The cell proliferation, alkaline phosphatase (ALP) activity, ALP stain, alizarin red stain, osteocalcin (OCN) protein expression, and osteogenic-specific gene expression were analyzed and compared among these groups. The outcome showed that all PRP lysate-treated groups had good ALP stain and ALP activity performance. Better alizarin red stains were found in the 2.5%, 5%, and 7.5% PRP lysate groups. The 2.5% and 5% PRP lysate groups showed superior results in OCN quantitative polymerase chain reaction, whereas the 5% and 7.5% PRP lysate groups showed higher OCN protein expressions. Early RUNX2 (Runt-related transcription factor 2 () genes were the most expressed in the 5% PRP lysate group, followed by the 2.5% PRP lysate group, and then the 7.5% PRP lysate group. Thus, we concluded that 5% PRP lysate seemed to provide the optimal effect on enhancing the osteogenic potential of ADSCs. Platelet-rich plasma lysate-treated ADSCs were considered to be a good cell source for application in treating nonunion or bone defects in the future.
Collapse
Affiliation(s)
- Chia-Fang Chen
- Division of Trauma Plastic Surgery, Department of Plastic and Reconstructive Surgery
| | | | | |
Collapse
|
7
|
Mu Y, Du Z, Gao W, Xiao L, Crawford R, Xiao Y. The effect of a bionic bone ionic environment on osteogenesis, osteoimmunology, and in situ bone tissue engineering. Biomaterials 2024; 304:122410. [PMID: 38043465 DOI: 10.1016/j.biomaterials.2023.122410] [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: 07/11/2023] [Revised: 11/15/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
Bone, a mineralized tissue, continuously undergoes remodeling. It is a process that engages the mineralization and demineralization of the bone matrix, orchestrated by the interactions among cells and cell-secreted biomolecules under the bone ionic microenvironment (BIE). The osteoinductive properties of the demineralized organic bone matrix and many biological factors have been well-investigated. However, the impact of the bone ionic environment on cell differentiation and osteogenesis remains largely unknown. In this study, we extracted and isolated inorganic bone components (bone-derived monetite, BM) using a low-temperature method and, for the first time, investigated whether the BIE could actively affect cell differentiation and regulate osteoimmune reactions. It was evidenced that the BIE could foster the osteogenesis of human bone marrow stromal cells (hBMSCs) and promote hBMSCs mineralization without using osteogenic inductive agents. Interestingly, it was noted that BIE resulted in intracellular mineralization, evidenced by intracellular accumulation of carbonate hydroxyapatite similar to that oberved in osteoblasts cultured in osteoinductive media. Additionally, BIE was found to enhance osteogenesis by generating a favorable osteoimmune environment. In a rat calvarial bone defect model, the osteogenic capacity of BIE was evaluated using a collagen type I-impregnated BM (Col-BM) composite. It showed that Col-BM significantly promoted new bone formation in the critical-size bone defect areas. Taken together, this is the first study that investigated the influence of the BIE on osteogenesis, osteoimmunology, and in situ bone tissue engineering. The innate osteoinductive potential of inorganic bone components, both in vitro and in vivo, not only expands the understanding of the BIE on osteogenesis but also benefits future biomaterials engineering for bone tissue regeneration.
Collapse
Affiliation(s)
- Yuqing Mu
- School of Medicine and Dentistry, Griffith University (GU), Gold Coast, QLD, 4222, Australia; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia; School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Zhibin Du
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia; School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Wendong Gao
- School of Medicine and Dentistry, Griffith University (GU), Gold Coast, QLD, 4222, Australia; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia; School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Lan Xiao
- School of Medicine and Dentistry, Griffith University (GU), Gold Coast, QLD, 4222, Australia; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia; School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Ross Crawford
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia; School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Yin Xiao
- School of Medicine and Dentistry, Griffith University (GU), Gold Coast, QLD, 4222, Australia; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia; School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia.
| |
Collapse
|
8
|
Choi J, Lee H. NFIB-MLL1 complex is required for the stemness and Dlx5-dependent osteogenic differentiation of C3H10T1/2 mesenchymal stem cells. J Biol Chem 2023; 299:105193. [PMID: 37633334 PMCID: PMC10519831 DOI: 10.1016/j.jbc.2023.105193] [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: 04/10/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 08/28/2023] Open
Abstract
Despite significant progress in our understanding of the molecular mechanism of mesenchymal stem cell (MSC) differentiation, less is known about the factors maintaining the stemness and plasticity of MSCs. Here, we show that the NFIB-MLL1 complex plays key roles in osteogenic differentiation and stemness of C3H10T1/2 MSCs. We find that depletion of either NFIB or MLL1 results in a severely hampered osteogenic potential and failed activation of key osteogenic transcription factors, such as Dlx5, Runx2, and Osx, following osteogenic stimuli. In addition, the NFIB-MLL1 complex binds directly to the promoter of Dlx5, and exogenous expression of Myc-Dlx5, but not the activation of either the BMP- or the Wnt-signaling pathway, is sufficient to restore the osteogenic potential of cells depleted of NFIB or MLL1. Moreover, chromatin immunoprecipitation (ChIP) and ChIP-sequencing analysis showed that the NFIB-MLL1 complex mediates the deposition of trimethylated histone H3K4 at both Dlx5 and Cebpa, key regulator genes that function at the early stages of osteogenic and adipogenic differentiation, respectively, in uncommitted C3H10T1/2 MSCs. Surprisingly, the depletion of either NFIB or MLL1 leads to decreased trimethylated histone H3K4 and results in elevated trimethylated histone H3K9 at those developmental genes. Furthermore, gene expression profiling and ChIP-sequencing analysis revealed lineage-specific changes in chromatin landscape and gene expression in response to osteogenic stimuli. Taken together, these data provide evidence for the hitherto unknown role of the NFIB-MLL1 complex in the maintenance and lineage-specific differentiation of C3H10T1/2 MSCs and support the epigenetic regulatory mechanism underlying the stemness and plasticity of MSCs.
Collapse
Affiliation(s)
- Janghyun Choi
- Department of Biological Sciences, College of Natural Science, Inha University, Incheon, South Korea.
| | - Hansol Lee
- Department of Biological Sciences, College of Natural Science, Inha University, Incheon, South Korea.
| |
Collapse
|
9
|
Kim KM, Son HE, Lim YJ, Jang WG. Topiramate promotes osteogenic differentiation through AMPK-dependent phosphorylation of Smad1/5/9. Acta Histochem 2023; 125:152095. [PMID: 37757516 DOI: 10.1016/j.acthis.2023.152095] [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: 07/21/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 09/29/2023]
Abstract
Topiramate [2,3:4,5-bis-o-(1-methylethylidene) β-D-fructo-pyranose sulfamate; TPM] is one of the most used new-generation antiepileptic drugs. It has been reported to regulate the differentiation of human bone cells. However, the molecular mechanism of TPM in osteoblast differentiation is not fully elucidated. In the present study, we examined the effect of TPM on osteogenic differentiation of C3H10T1/2, MC3T3-E1, primary mouse calvarial cells, and primary bone marrow stem cells (BMSCs). Primary cells were isolated from mice calvaria and bone marrow respectively. Expression of the osteogenic gene was determined by RT-PCR. The osteogenic protein levels were measured by Western blot analysis. Alkaline phosphatase (ALP) staining experiment was performed to evaluate ALP activity. Alizarin red s (ARS) staining was performed to measure zebrafish caudal fin regeneration. Treatment of TPM up-regulated the osteogenic genes including distal-less homeobox 5 (Dlx5) and runt-related transcription factor 2 (Runx2). In addition, TPM also increased the Dlx5 and Runx2 protein levels, Smad1/5/9 phosphorylation, and alkaline phosphatase (ALP) activity. Furthermore, TPM activated AMPK, and inhibition of AMPK decreased TPM-induced osteogenic differentiation. In the zebrafish model, osteogenic effect of TPM was identified. TPM was increased amputated caudal fin rays of zebrafish. These results demonstrate that TPM enhances osteogenic differentiation via AMPK-mediated Smad1/5/9 phosphorylation.
Collapse
Affiliation(s)
- Kyeong-Min Kim
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk 38453, Republic of Korea; Research Institute of Anti-Aging, Daegu University, Gyeongbuk 38453, Republic of Korea
| | - Hyo-Eun Son
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk 38453, Republic of Korea; Research Institute of Anti-Aging, Daegu University, Gyeongbuk 38453, Republic of Korea
| | - Young-Ju Lim
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk 38453, Republic of Korea; Research Institute of Anti-Aging, Daegu University, Gyeongbuk 38453, Republic of Korea
| | - Won-Gu Jang
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk 38453, Republic of Korea; Research Institute of Anti-Aging, Daegu University, Gyeongbuk 38453, Republic of Korea.
| |
Collapse
|
10
|
Fujii S, Takebe H, Mizoguchi T, Nakamura H, Shimo T, Hosoya A. Bone formation ability of Gli1 + cells in the periodontal ligament after tooth extraction. Bone 2023; 173:116786. [PMID: 37164217 DOI: 10.1016/j.bone.2023.116786] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/29/2023] [Accepted: 04/30/2023] [Indexed: 05/12/2023]
Abstract
During the process of socket healing after tooth extraction, osteoblasts appear in the tooth socket and form alveolar bone; however, the source of these osteoblasts is still uncertain. Recently, it has been demonstrated that cells expressing Gli1, a downstream factor of sonic hedgehog signaling, exhibit stem cell properties in the periodontal ligament (PDL). Therefore, in the present study, the differentiation ability of Gli1+-PDL cells after tooth extraction was analyzed using Gli1-CreERT2/ROSA26-loxP-stop-loxP-tdTomato (iGli1/Tomato) mice. After the final administration of tamoxifen to iGli1/Tomato mice, Gli1/Tomato+ cells were rarely detected in the PDL. One day after the tooth extraction, although inflammatory cells appeared in the tooth socket, Periostin+ PDL-like tissues having a few Gli1/Tomato+ cells remained near the alveolar bone. Three days after the extraction, the number of Gli1/Tomato+ cells increased as evidenced by numerous PCNA+ cells in the socket. Some of these Gli1/Tomato+ cells expressed BMP4 and Phosphorylated (P)-Smad1/5/8. After seven days, the Osteopontin+ bone matrix was formed in the tooth socket apart from the alveolar bone. Many Gli1/Tomato+ osteoblasts that were positive for Runx2+ were arranged on the surface of the newly formed bone matrix. In the absence of Gli1+-PDL cells in Gli1-CreERT2/Rosa26-loxP-stop-loxP-tdDTA (iGli1/DTA) mice, the amount of newly formed bone matrix was significantly reduced in the tooth socket. Therefore, these results collectively suggest that Gli1+-PDL cells differentiate into osteoblasts to form the bone matrix in the tooth socket; thus, this differentiation might be regulated, at least in part, by bone morphogenetic protein (BMP) signaling.
Collapse
Affiliation(s)
- Saki Fujii
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan; Division of Reconstructive Surgery for Oral and Maxillofacial Region, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| | - Hiroaki Takebe
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| | | | - Hiroaki Nakamura
- Department of Oral Anatomy, Matsumoto Dental University, Nagano, Japan
| | - Tsuyoshi Shimo
- Division of Reconstructive Surgery for Oral and Maxillofacial Region, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| | - Akihiro Hosoya
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan.
| |
Collapse
|
11
|
Hisaoka S, Osawa J, Kobashi R, Ishida A, Kameshita I, Sueyoshi N. Subcellular distribution of bone morphogenetic protein 2-inducible kinase (BMP2K): Regulation by liquid-liquid phase separation and nucleocytoplasmic shuttling. Biochem Biophys Res Commun 2023; 649:16-24. [PMID: 36739695 DOI: 10.1016/j.bbrc.2023.01.076] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/12/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
Bone morphogenetic protein 2 (BMP2)-inducible kinase (BMP2K) is induced by the cytokine BMP2, which is also implicated in the production of bone differentiation. In addition to regulating bone differentiation, BMP2K is implicated in a variety of cancers. Therefore, understanding the variables that determine where in the cell this kinase functions may help in understanding malignancies linked to BMP2K. However, the mechanisms regulating the subcellular localization of BMP2K are mainly unknown. By liquid-liquid phase separation (LLPS), BMP2K forms droplets in the cytoplasm, but how the droplets are regulated remains unclear. The reason why BMP2K localizes to the cytoplasm irrespective of having a nuclear localization signal (NLS) is also unknown. Here we show the element that controls BMP2K's LLPS and cytoplasmic localization. A glutamine-rich area is necessary for BMP2K phase separation, and droplet formation is controlled by hyperosmolarity. Cytoplasmic localization of BMP2K is managed by inhibition of NLS function through phosphorylation of Ser-1010 and by a newly found cytoplasmic localization region that antagonizes the NLS. These results will provide an important biochemical foundation for the advancement of BMP2K-related cell biology, structural biology, and pathophysiology.
Collapse
Affiliation(s)
- Shiho Hisaoka
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa, 761-0795, Japan
| | - Jin Osawa
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa, 761-0795, Japan
| | - Riku Kobashi
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa, 761-0795, Japan
| | - Atsuhiko Ishida
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima, 739-8521, Japan.
| | - Isamu Kameshita
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa, 761-0795, Japan
| | - Noriyuki Sueyoshi
- Department of Life Sciences, Faculty of Agriculture, Kagawa University, Kagawa, 761-0795, Japan.
| |
Collapse
|
12
|
Sim Y, Seo HJ, Kim DH, Lee SH, Kwon J, Kwun IS, Jung C, Kim JI, Lim JH, Kim DK, Baek MC, Cho YE. The Effect of Apple-Derived Nanovesicles on the Osteoblastogenesis of Osteoblastic MC3T3-E1 Cells. J Med Food 2023; 26:49-58. [PMID: 36594993 DOI: 10.1089/jmf.2022.k.0094] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Osteoporosis is characterized by low bone mass and elevated structural deterioration of the bone tissue, resulting in bone weakness with an increased risk of fracture. Considering biological activities of various phytochemicals extracted from apples, we herein demonstrated the potential antiosteoporotic effects of apple-derived nanovesicles (apple NVs) using osteoblastic MC3T3-E1 cells. Apple NVs significantly stimulated the growth of MC3T3-E1 cells. The cellular alkaline phosphatase (ALP) activity was significantly upregulated in the 5 μg/mL apple NVs-treated group. In addition, the concentrarion of mineralized nodules was significantly increased in the apple NVs-treated groups. Furthermore, apple NVs increased the expression of the genes and proteins associated with osteoblast growth and differentiation, such as Runx2, ALP, OPN, and BMP2/4, which further activated ERK- and JNK-related mitogen-activated protein kinase signaling. These results demonstrate that apple NVs have a potential to prevent osteoporosis by promoting osteoblastogenesis in osteoblastic MC3T3-E1 cells through regulating the BMP2/Smad1 pathways.
Collapse
Affiliation(s)
- Yejin Sim
- Department of Food and Nutrition, Andong National University, Andong, South Korea
| | - Hyun-Ju Seo
- Department of Food and Nutrition, Andong National University, Andong, South Korea
- Department of Molecular Medicine, CMRI, School of Medicine, Kyungpook National University, Daegu, South Korea
- Agriculture Science and Technology Research Institute, Andong National University, Andong, South Korea
| | - Dong-Ha Kim
- Department of Molecular Medicine, CMRI, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Sang-Hoon Lee
- Department of Food and Nutrition, Andong National University, Andong, South Korea
| | - JaeHee Kwon
- Department of Food and Nutrition, Andong National University, Andong, South Korea
| | - In-Sook Kwun
- Department of Food and Nutrition, Andong National University, Andong, South Korea
| | - Chuleui Jung
- Agriculture Science and Technology Research Institute, Andong National University, Andong, South Korea
- Department of Plant Medicals, Andong National University, Andong, South Korea
| | - Jee-In Kim
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Jae-Hwan Lim
- Department of Biological Science, Andong National University, Andong, South Korea
- Institute of Vaccine Biotechnology, Andong National University, Andong, South Korea
| | - Do-Kyun Kim
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeonbuk, South Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Young-Eun Cho
- Department of Food and Nutrition, Andong National University, Andong, South Korea
- Institute of Vaccine Biotechnology, Andong National University, Andong, South Korea
| |
Collapse
|
13
|
In Vitro and In Vivo Studies of Hydrogenated Titanium Dioxide Nanotubes with Superhydrophilic Surfaces during Early Osseointegration. Cells 2022; 11:cells11213417. [DOI: 10.3390/cells11213417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/23/2022] [Accepted: 10/26/2022] [Indexed: 11/17/2022] Open
Abstract
Titanium-based implants are often utilized in oral implantology and craniofacial reconstructions. However, the biological inertness of machined titanium commonly results in unsatisfactory osseointegration. To improve the osseointegration properties, we modified the titanium implants with nanotubular/superhydrophilic surfaces through anodic oxidation and thermal hydrogenation and evaluated the effects of the machined surfaces (M), nanotubular surfaces (Nano), and hydrogenated nanotubes (H-Nano) on osteogenesis and osseointegration in vitro and in vivo. After incubation of mouse bone marrow mesenchymal stem cells on the samples, we observed improved cell adhesion, alkaline phosphatase activity, osteogenesis-related gene expression, and extracellular matrix mineralization in the H-Nano group compared to the other groups. Subsequent in vivo studies indicated that H-Nano implants promoted rapid new bone regeneration and osseointegration at 4 weeks, which may be attributed to the active osteoblasts adhering to the nanotubular/superhydrophilic surfaces. Additionally, the Nano group displayed enhanced osteogenesis in vitro and in vivo at later stages, especially at 8 weeks. Therefore, we report that hydrogenated superhydrophilic nanotubes can significantly accelerate osteogenesis and osseointegration at an early stage, revealing the considerable potential of this implant modification for clinical applications.
Collapse
|
14
|
Hang R, Wang J, Tian X, Wu R, Hang R, Zhao Y, Sun Y, Wang H. Resveratrol promotes osteogenesis and angiogenesis through mediating immunology of senescent macrophages. Biomed Mater 2022; 17. [PMID: 35830846 DOI: 10.1088/1748-605x/ac80e3] [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: 04/23/2022] [Accepted: 07/13/2022] [Indexed: 11/12/2022]
Abstract
Orthopedic implants have been used clinically to restore the functions of the compromised bone tissues, but there is still a relatively high risk of failure for elderly people. A critical reason is pro-inflammatory immune microenvironment created by senescent macrophages with homeostasis imbalance impairs osteogenesis and angiogenesis, two major processes involved in implant osseointegration. The present work proposes to use resveratrol as an autophagy inducing agent to upregulate the autophagy level of senescent macrophages to restore homeostasis, consequently generating a favorable immune microenvironment. The results show 0.1-1 µM of resveratrol can induce autophagy of senescent macrophages, promote cell viability and proliferation, reduce intracellular reactive oxygen species (ROS) level, and polarize the cells to pro-healing M2 phenotype. The immune microenvironment created by senescent macrophages upon resveratrol stimulation can promote osteogenesis and angiogenesis, as manifested by upregulated proliferation, alkaline phosphatase activity, type I collagen secretion, and extracellular matrix mineralization of senescent osteoblasts as well as nitric oxide production, migration, and in vitro angiogenesis of senescent endothelial cells. In addition, resveratrol-loaded silk fibroin coatings can be fabricated on titanium surface through electrophoretic co-deposition and the coatings show beneficial effects on the functions of senescent macrophages. Our results suggest resveratrol can be used as surface additive of titanium implants to promote osseointegration of elderly people though regulating immunology of senescent macrophages.
Collapse
Affiliation(s)
- Ruiqiang Hang
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, Shanxi , 030024, CHINA
| | - Jiahui Wang
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Xue Tian
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Ruifeng Wu
- Central Hospital of Tongchuan Mining Bureau, No. 15, Chuankou Road, Tongchuan, Shaanxi Province, Tongchuan, 727000, CHINA
| | - Ruiyue Hang
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Yuyu Zhao
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Yonghua Sun
- Taiyuan University of Technology, No. 79, Yingze West Road, Taiyuan, Shanxi Province, Taiyuan, 030024, CHINA
| | - Honggang Wang
- Central Hospital of Tongchuan Mining Bureau, No. 15, Chuankou Road, Tongchuan, Shaanxi Province, Taiyuan, 030024, CHINA
| |
Collapse
|
15
|
Poleboina S, Sheth VG, Sharma N, Sihota P, Kumar N, Tikoo K. Selenium nanoparticles stimulate osteoblast differentiation via BMP-2/MAPKs/β-catenin pathway in diabetic osteoporosis. Nanomedicine (Lond) 2022; 17:607-625. [DOI: 10.2217/nnm-2021-0401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To evaluate whether selenium nanoparticles (SeNPs) can stimulate bone formation and inhibit the bone loss involved in hyperglycemia-induced osteoporosis. Methods: Rat osteoblastic UMR-106 cells were used for in vitro studies and female Sprague–Dawley rats were used for type 2 diabetes-associated osteoporosis in vivo study. Results: In vitro studies show that SeNPs promote osteoblast differentiation via modulating alkaline phosphatase (ALP) activity, and promoting calcium nodule formation and collagen content. The authors also provide evidence regarding the involvement of the BMP-2/MAPKs/β-catenin pathway in preventing diabetic osteoporosis. Further, in vivo and ex vivo studies suggested that SeNPs can preserve mechanical and microstructural properties of bone. Conclusion: To the best of our knowledge, this study provides the first evidence regarding the therapeutic benefits of SeNPs in preventing diabetes-associated bone fragility.
Collapse
Affiliation(s)
- Sumathi Poleboina
- Department of Pharmacology & Toxicology, Laboratory of Epigenetics & Diseases, National Institute of Pharmaceutical Education & Research, Sector-67, S.A.S. Nagar, Punjab, 160062, India
| | - Vaibhav G Sheth
- Department of Pharmacology & Toxicology, Laboratory of Epigenetics & Diseases, National Institute of Pharmaceutical Education & Research, Sector-67, S.A.S. Nagar, Punjab, 160062, India
| | - Nisha Sharma
- Department of Pharmacology & Toxicology, Laboratory of Epigenetics & Diseases, National Institute of Pharmaceutical Education & Research, Sector-67, S.A.S. Nagar, Punjab, 160062, India
| | - Praveer Sihota
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, 14000, India
| | - Navin Kumar
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, 14000, India
| | - Kulbhushan Tikoo
- Department of Pharmacology & Toxicology, Laboratory of Epigenetics & Diseases, National Institute of Pharmaceutical Education & Research, Sector-67, S.A.S. Nagar, Punjab, 160062, India
| |
Collapse
|
16
|
BMP-2 Enhances Osteogenic Differentiation of Human Adipose-Derived and Dental Pulp Stem Cells in 2D and 3D In Vitro Models. Stem Cells Int 2022; 2022:4910399. [PMID: 35283997 PMCID: PMC8916887 DOI: 10.1155/2022/4910399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/02/2021] [Accepted: 11/08/2021] [Indexed: 12/25/2022] Open
Abstract
Bone tissue provides support and protection to different organs and tissues. Aging and different diseases can cause a decrease in the rate of bone regeneration or incomplete healing; thus, tissue-engineered substitutes can be an acceptable alternative to traditional therapies. In the present work, we have developed an in vitro osteogenic differentiation model based on mesenchymal stem cells (MSCs), to first analyse the influence of the culture media and the origin of the cells on the efficiency of this process and secondly to extrapolate it to a 3D environment to evaluate its possible application in bone regeneration therapies. Two osteogenic culture media were used (one commercial from Stemcell Technologies and a second supplemented with dexamethasone, ascorbic acid, glycerol-2-phosphate, and BMP-2), with human cells of a mesenchymal phenotype from two different origins: adipose tissue (hADSCs) and dental pulp (hDPSCs). The expression of osteogenic markers in 2D cultures was evaluated in several culture periods by means of the immunofluorescence technique and real-time gene expression analysis, taking as reference MG-63 cells of osteogenic origin. The same strategy was extrapolated to a 3D environment of polylactic acid (PLA), with a 3% alginate hydrogel. The expression of osteogenic markers was detected in both hADSCs and hDPSCs, cultured in either 2D or 3D environments. However, the osteogenic differentiation of MSCs was obtained based on the culture medium and the cell origin used, since higher osteogenic marker levels were found when hADSCs were cultured with medium supplemented with BMP-2. Furthermore, the 3D culture used was suitable for cell survival and osteogenic induction.
Collapse
|
17
|
lncRNAs MALAT1 and LINC00657 upstream to miR-214-3p/BMP2 regulate osteogenic differentiation of human mesenchymal stem cells. Mol Biol Rep 2022; 49:6847-6857. [DOI: 10.1007/s11033-022-07136-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 01/10/2022] [Indexed: 10/19/2022]
|
18
|
Injectable bioactive polymethyl methacrylate–hydrogel hybrid bone cement loaded with BMP-2 to improve osteogenesis for percutaneous vertebroplasty and kyphoplasty. Biodes Manuf 2022. [DOI: 10.1007/s42242-021-00172-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
19
|
Kang F, Yi Q, Gu P, Dong Y, Zhang Z, Zhang L, Bai Y. Controlled growth factor delivery system with osteogenic-angiogenic coupling effect for bone regeneration. J Orthop Translat 2022; 31:110-125. [PMID: 34976731 PMCID: PMC8671819 DOI: 10.1016/j.jot.2021.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/01/2021] [Accepted: 11/15/2021] [Indexed: 11/19/2022] Open
Abstract
Objective Bone regeneration involves a coordinated cascade of events that are regulated by several cytokines and growth factors, among which bone morphogenic protein-2 (BMP-2), vascular endothelial growth factor (VEGF) and fibroblast growth factor-2 (FGF-2) play important roles. In this study, we investigated the effects of dual release of the three growth factors on bone regeneration in femur defects. Methods A composite consisting of Gelatin microparticles loaded with VEGF/FGF-2 and poly(lactic-co-glycolic acid)-poly(ethylene glycol)-carboxyl (PLGA-PEG-COOH) microparticles loaded with BMP-2 encapsulated in a nano hydroxyapatite-poly actic-co-glycolic acid (nHA-PLGA) scaffold was prepared for the dual release of the growth factors. Results On the 14th day, decreased release rate of BMP-2 compared with FGF-2 and VEGF was observed. However, after 14 days, compared to FGF-2 and VEGF, BMP-2 showed an increased release rate. Controlled dual release of BMP-2 and VEGF, FGF-2 resulted in a significant osteogenic differentiation of bone mesenchymal stem cells (BMSCs). Moreover, effects of the composite scaffold on functional connection of osteoblast-vascular cells during bone development were evaluated. The synergistic effects of dual delivery of growth factors were shown to promote the expression of VEGF in BMSCs. Increased secretion of VEGF from BMSCs promoted the proliferation and angiogenic differentiation of human umbilical vein endothelial cells (HUVECs) in the co-culture system. At 12 weeks after implantation, blood vessel and bone formation were analyzed by micro-CT and histology. The composite scaffold significantly promoted the formation of blood vessels and new bone in femur defects. Conclusions These findings demonstrate that dual delivery of angiogenic factors and osteogenic factors from Gelatin and PLGA-PEG-COOH microparticles-based composite scaffolds exerted an osteogenic-angiogenic coupling effect on bone regeneration. This approach will inform on the development of appropriate designs of high-performance bioscaffolds for bone tissue engineering.
Collapse
Affiliation(s)
- Fei Kang
- Department of Biomedical Materials Science, Third Military Medical University, Chongqing, 400038, China
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Qiying Yi
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Pengcheng Gu
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yuhan Dong
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Ziyang Zhang
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Lijuan Zhang
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yan Bai
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- Corresponding author. School of Pharmacy, Chongqing Medical University, District of Yuzhong, Chongqing, 400016, PR China.
| |
Collapse
|
20
|
Wang M, Li W, Mille LS, Ching T, Luo Z, Tang G, Garciamendez CE, Lesha A, Hashimoto M, Zhang YS. Digital Light Processing Based Bioprinting with Composable Gradients. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107038. [PMID: 34609032 PMCID: PMC8741743 DOI: 10.1002/adma.202107038] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Indexed: 05/23/2023]
Abstract
Recapitulation of complex tissues signifies a remarkable challenge and, to date, only a few approaches have emerged that can efficiently reconstruct necessary gradients in 3D constructs. This is true even though mimicry of these gradients is of great importance to establish the functionality of engineered tissues and devices. Here, a composable-gradient Digital Light Processing (DLP)-based (bio)printing system is developed, utilizing the unprecedented integration of a microfluidic mixer for the generation of either continual or discrete gradients of desired (bio)inks in real time. Notably, the precisely controlled gradients are composable on-the-fly by facilely by adjusting the (bio)ink flow ratios. In addition, this setup is designed in such a way that (bio)ink waste is minimized when exchanging the gradient (bio)inks, further enhancing this time- and (bio)ink-saving strategy. Various planar and 3D structures exhibiting continual gradients of materials, of cell densities, of growth factor concentrations, of hydrogel stiffness, and of porosities in horizontal and/or vertical direction, are exemplified. The composable fabrication of multifunctional gradients strongly supports the potential of the unique bioprinting system in numerous biomedical applications.
Collapse
Affiliation(s)
- Mian Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Wanlu Li
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Luis S. Mille
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Terry Ching
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore 48737
- Digital Manufacturing and Design Centre, Singapore University of Technology and Design, Singapore 487372
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583
| | - Zeyu Luo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Guosheng Tang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Carlos Ezio Garciamendez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Ami Lesha
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Michinao Hashimoto
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore 48737
- Digital Manufacturing and Design Centre, Singapore University of Technology and Design, Singapore 487372
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
21
|
Papanota AM, Karousi P, Kontos CK, Artemaki PI, Liacos CI, Papadimitriou MA, Bagratuni T, Eleutherakis-Papaiakovou E, Malandrakis P, Ntanasis-Stathopoulos I, Gavriatopoulou M, Kastritis E, Avgeris M, Dimopoulos MA, Scorilas A, Terpos E. A Cancer-Related microRNA Signature Shows Biomarker Utility in Multiple Myeloma. Int J Mol Sci 2021; 22:13144. [PMID: 34884950 PMCID: PMC8658678 DOI: 10.3390/ijms222313144] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy, arising from terminally differentiated B cells, namely plasma cells. miRNAs are small non-coding RNAs that participate in the post-transcriptional regulation of gene expression. In this study, we investigated the role of nine miRNAs in MM. CD138+ plasma cells were selected from bone marrow aspirates from MM and smoldering MM (sMM) patients. Total RNA was extracted and in vitro polyadenylated. Next, first-strand cDNA synthesis was performed using an oligo-dT-adapter primer. For the relative quantification of the investigated miRNAs, an in-house real-time quantitative PCR (qPCR) assay was developed. A functional in silico analysis of the miRNAs was also performed. miR-16-5p and miR-155-5p expression was significantly lower in the CD138+ plasma cells of MM patients than in those of sMM patients. Furthermore, lower levels of miR-15a-5p, miR-16-5p, and miR-222-3p were observed in the CD138+ plasma cells of MM patients with osteolytic bone lesions, compared to those without. miR-125b-5p was also overexpressed in the CD138+ plasma cells of MM patients with bone disease that presented with skeletal-related events (SREs). Furthermore, lower levels of miR-223-3p were associated with significantly worse overall survival in MM patients. In conclusion, we propose a miRNA signature with putative clinical utility in MM.
Collapse
Affiliation(s)
- Aristea-Maria Papanota
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-M.P.); (C.-I.L.); (T.B.); (E.E.-P.); (P.M.); (I.N.-S.); (M.G.); (E.K.); (M.-A.D.)
| | - Paraskevi Karousi
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece; (P.K.); (C.K.K.); (P.I.A.); (M.-A.P.); (M.A.)
| | - Christos K. Kontos
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece; (P.K.); (C.K.K.); (P.I.A.); (M.-A.P.); (M.A.)
| | - Pinelopi I. Artemaki
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece; (P.K.); (C.K.K.); (P.I.A.); (M.-A.P.); (M.A.)
| | - Christine-Ivy Liacos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-M.P.); (C.-I.L.); (T.B.); (E.E.-P.); (P.M.); (I.N.-S.); (M.G.); (E.K.); (M.-A.D.)
| | - Maria-Alexandra Papadimitriou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece; (P.K.); (C.K.K.); (P.I.A.); (M.-A.P.); (M.A.)
| | - Tina Bagratuni
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-M.P.); (C.-I.L.); (T.B.); (E.E.-P.); (P.M.); (I.N.-S.); (M.G.); (E.K.); (M.-A.D.)
| | - Evangelos Eleutherakis-Papaiakovou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-M.P.); (C.-I.L.); (T.B.); (E.E.-P.); (P.M.); (I.N.-S.); (M.G.); (E.K.); (M.-A.D.)
| | - Panagiotis Malandrakis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-M.P.); (C.-I.L.); (T.B.); (E.E.-P.); (P.M.); (I.N.-S.); (M.G.); (E.K.); (M.-A.D.)
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-M.P.); (C.-I.L.); (T.B.); (E.E.-P.); (P.M.); (I.N.-S.); (M.G.); (E.K.); (M.-A.D.)
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-M.P.); (C.-I.L.); (T.B.); (E.E.-P.); (P.M.); (I.N.-S.); (M.G.); (E.K.); (M.-A.D.)
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-M.P.); (C.-I.L.); (T.B.); (E.E.-P.); (P.M.); (I.N.-S.); (M.G.); (E.K.); (M.-A.D.)
| | - Margaritis Avgeris
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece; (P.K.); (C.K.K.); (P.I.A.); (M.-A.P.); (M.A.)
- Laboratory of Clinical Biochemistry-Molecular Diagnostics, Second Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “P. & A. Kyriakou” Children’s Hospital, 11527 Athens, Greece
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-M.P.); (C.-I.L.); (T.B.); (E.E.-P.); (P.M.); (I.N.-S.); (M.G.); (E.K.); (M.-A.D.)
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece; (P.K.); (C.K.K.); (P.I.A.); (M.-A.P.); (M.A.)
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-M.P.); (C.-I.L.); (T.B.); (E.E.-P.); (P.M.); (I.N.-S.); (M.G.); (E.K.); (M.-A.D.)
| |
Collapse
|
22
|
Li C, Fennessy P. The periosteum: a simple tissue with many faces, with special reference to the antler-lineage periostea. Biol Direct 2021; 16:17. [PMID: 34663443 PMCID: PMC8522104 DOI: 10.1186/s13062-021-00310-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 10/12/2021] [Indexed: 11/30/2022] Open
Abstract
Periosteum is a thin membrane covering bone surfaces and consists of two layers: outer fibrous layer and inner cambium layer. Simple appearance of periosteum has belied its own complexity as a composite structure for physical bone protection, mechano-sensor for sensing mechanical loading, reservoir of biochemical molecules for initiating cascade signaling, niche of osteogenic cells for bone formation and repair, and "umbilical cord" for nourishing bone tissue. Periosteum-derived cells (PDCs) have stem cell attributes: self-renewal (no signs of senescence until 80 population doublings) and multipotency (differentiate into fibroblasts, osteoblasts, chondrocytes, adipocytes and skeletal myocytes). In this review, we summarized the currently available knowledge about periosteum and with special references to antler-lineage periostea, and demonstrated that although periosteum is a type of simple tissue in appearance, with multiple faces in functions; antler-lineage periostea add another dimension to the properties of somatic periostea: capable of initiation of ectopic organ formation upon transplantation and full mammalian organ regeneration when interacted with the covering skin. Very recently, we have translated this finding into other mammals, i.e. successfully induced partial regeneration of the amputated rat legs. We believe further refinement along this line would greatly benefit human health.
Collapse
Affiliation(s)
- Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, 1345 Pudong Rd., Changchun, 130000, Jilin, China.
| | - Peter Fennessy
- AbacusBio Limited, 442 Moray Place, Dunedin, New Zealand
| |
Collapse
|
23
|
Kirankumar S, Gurusamy N, Rajasingh S, Sigamani V, Vasanthan J, Perales SG, Rajasingh J. Modern approaches on stem cells and scaffolding technology for osteogenic differentiation and regeneration. Exp Biol Med (Maywood) 2021; 247:433-445. [PMID: 34648374 DOI: 10.1177/15353702211052927] [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] [Indexed: 12/11/2022] Open
Abstract
The process of bone repair has always been a natural mystery. Although bones do repair themselves, supplemental treatment is required for the initiation of the self-regeneration process. Predominantly, surgical procedures are employed for bone regeneration. Recently, cell-based therapy for bone regeneration has proven to be more effective than traditional methods, as it eliminates the immune risk and painful surgeries. In clinical trials, various stem cells, especially mesenchymal stem cells, have shown to be more efficient for the treatment of several bone-related diseases, such as non-union fracture, osteogenesis imperfecta, osteosarcoma, and osteoporosis. Furthermore, the stem cells grown in a suitable three-dimensional scaffold support were found to be more efficient for osteogenesis. It has been shown that the three-dimensional bioscaffolds support and simulate an in vivo environment, which helps in differentiation of stem cells into bone cells. Bone regeneration in patients with bone disorders can be improved through modification of stem cells with several osteogenic factors or using stem cells as carriers for osteogenic factors. In this review, we focused on the various types of stem cells and scaffolds that are being used for bone regeneration. In addition, the molecular mechanisms of various transcription factors, signaling pathways that support bone regeneration and the senescence of the stem cells, which limits bone regeneration, have been discussed.
Collapse
Affiliation(s)
- Shivaani Kirankumar
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Genetic Engineering, 93104SRM Institute of Science and Technology, Chennai 603203, India
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jayavardini Vasanthan
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Genetic Engineering, 93104SRM Institute of Science and Technology, Chennai 603203, India
| | - Selene G Perales
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
24
|
Mantsou A, Pitou M, Papachristou E, Papi RM, Lamprou P, Choli-Papadopoulou T. Effect of a Bone Morphogenetic Protein-2-derived peptide on the expression of tumor marker ZNF217 in osteoblasts and MCF-7 cells. Bone Rep 2021; 15:101125. [PMID: 34632002 PMCID: PMC8487976 DOI: 10.1016/j.bonr.2021.101125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/27/2021] [Accepted: 09/01/2021] [Indexed: 11/24/2022] Open
Abstract
Zinc Finger Protein 217 (ZNF217), a transcription factor and oncogene product, has been found to dysregulate Bone Morphogenetic Protein (BMP) signaling and induce invasion in breast tumors. In this study, the effect of BMP-2 or an active BMP-2 peptide, AISMLYLDEN, on the expression of ZNF217, BMP4 and CDK-inhibitor p21 gene, CDKN1A, was investigated in MCF-7 breast cancer cells. In parallel, the entire protein (BMP-2) as well as the aforementioned peptide were investigated in hDPSCs during osteogenic differentiation. The treatment of MCF-7 cancer cells with different concentrations of peptide AISMLYLDEN showed that the addition of 22.6 ng/ml was more effective in comparison to the other used concentrations. In particular, 48 h after treatment, CDKN1A and BMP4 mRNA levels were substantially increased in contrast to ZNF217 mRNA levels which were decreased. These results are strongly supported by BrdU assay that clearly indicated inhibition of cancer cell proliferation. Taken together, these results open ways for a concurrent use, at appropriate concentrations, of the peptide AISMLYLDEN during conventional therapeutic treatment in breast tumors with a metastatic tendency to the bones. Regarding the effect of the entire protein as well as its peptide on hDPSCs differentiation into osteocytes, the mRNA levels of osteocalcin, an osteogenic marker, showed that the peptide enhanced osteogenesis at a higher degree in comparison to the entire BMP-2 without however altering ZNF217, CDKN1A and BMP4 expression levels, which remained as expected of non-cancer cells.
Collapse
Affiliation(s)
- Aglaia Mantsou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Maria Pitou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Eleni Papachristou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Rigini M Papi
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Paraskevas Lamprou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Theodora Choli-Papadopoulou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| |
Collapse
|
25
|
Mollentze J, Durandt C, Pepper MS. An In Vitro and In Vivo Comparison of Osteogenic Differentiation of Human Mesenchymal Stromal/Stem Cells. Stem Cells Int 2021; 2021:9919361. [PMID: 34539793 PMCID: PMC8443361 DOI: 10.1155/2021/9919361] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/23/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
The use of stem cells in regenerative medicine, including tissue engineering and transplantation, has generated a great deal of enthusiasm. Mesenchymal stromal/stem cells (MSCs) can be isolated from various tissues, most commonly, bone marrow but more recently adipose tissue, dental pulp, and Wharton's jelly, to name a few. MSCs display varying phenotypic profiles and osteogenic differentiating capacity depending and their site of origin. MSCs have been successfully differentiated into osteoblasts both in vitro an in vivo but discrepancies exist when the two are compared: what happens in vitro does not necessarily happen in vivo, and it is therefore important to understand why these differences occur. The osteogenic process is a complex network of transcription factors, stimulators, inhibitors, proteins, etc., and in vivo experiments are helpful in evaluating the various aspects of this osteogenic process without distractions and confounding variables. With that in mind, the results of in vitro experiments need to be carefully considered and interpreted with caution as they do not perfectly replicate the conditions found within living organisms. This is where in vivo experiments help us better understand interactions that might occur in the osteogenic process that cannot be replicated in vitro. Potentially, these differences could also be exploited to develop an optimal MSC cell therapeutic product that can be used for bone disorders. There are many bone disorders, most of which cause a great deal of discomfort. Clinically acceptable protocols could be developed in which MSCs are used to aid in bone regeneration providing relief for patients with chronic pain. The aim of this review is to examine the differences between studies conducted in vitro and in vivo with regard to the osteogenic process to better define the gaps in current osteogenic research. By better understanding osteogenic differentiation, we can better define treatment strategies for various bone disorders.
Collapse
Affiliation(s)
- Jamie Mollentze
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Chrisna Durandt
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
26
|
Lui H, Samsonraj RM, Vaquette C, Denbeigh J, Kakar S, Cool SM, Dudakovic A, van Wijnen AJ. Combination of BMP2 and EZH2 Inhibition to Stimulate Osteogenesis in a 3D Bone Reconstruction Model. Tissue Eng Part A 2021; 27:1084-1098. [PMID: 33234056 PMCID: PMC8851245 DOI: 10.1089/ten.tea.2020.0218] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022] Open
Abstract
High concentrations of bone morphogenetic protein 2 (BMP2) in bone regeneration cause adverse events (e.g, heterotopic bone formation and acute inflammation). This study examines novel epigenetic strategies (i.e., EZH2 inhibition) for augmenting osteogenesis, thereby aiming to reduce the required BMP2 dose in vivo for bone regeneration and minimize these adverse effects. Human bone marrow-derived mesenchymal stem cells (BMSCs) were grown on three-dimensional (3D)-printed medical-grade polycaprolactone scaffolds and incubated in osteogenic media containing 50 ng/mL BMP2 and/or 5 μM GSK126 (EZH2 inhibitor) for 6 days (n = 3 per group and timepoint). Constructs were harvested for realtime quantitative polymerase chain reaction analysis at Day 10 and immunofluorescence (IF) microscopy at Day 21. After pretreating for 6 days and maintaining in osteogenic media for 4 days, BMSC-seeded scaffolds were also implanted in an immunocompromised subcutaneous murine model (n = 39; 3/group/donor and 3 control scaffolds) for histological analysis at 8 weeks. Pretreatment of BMSCs with BMP2 and BMP2/GSK126 costimulated expression of osteoblast-related genes (e.g., IBSP, SP7, RUNX2, and DLX5), as well as protein accumulation (e.g., collagen type 1/COL1A1 and osteocalcin/BGLAP) based on IF staining. While in vivo implantation for 8 weeks did not result in bone formation, increased angiogenesis was observed in BMP2 and BMP2/GSK126 groups. This study finds that BMP2 and GSK126 costimulate osteogenic differentiation of MSCs on 3D scaffolds in vitro and may contribute to enhanced vascularization when implanted in vivo to support bone formation. Thus, epigenetic priming with EZH2 inhibitors may have translational potential in bone healing by permitting a reduction of BMP2 dosing in vivo to mitigate its side effects. Impact statement While autografts are still the gold standard for bone reconstruction, tissue availability and donor morbidity are significant limitations. Previous attempts to use high concentrations of bone morphogenetic protein 2 (BMP2) have been shown to cause adverse events such as excessive bone formation and acute inflammation. Overall, the utilization of EZH2 inhibitors to modulate gene expression in favor of bone healing has been demonstrated in vitro in a tissue engineering strategy. Our study will pave the way to developing tissue engineering strategies involving GSK126 as an adjuvant to increase the effects of BMP2 for stimulating cells of interest on a three-dimensional scaffold for bone regeneration.
Collapse
Affiliation(s)
- Hayman Lui
- School of Medicine, Griffith University, Gold Coast, Queensland, Australia
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Rebekah M. Samsonraj
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Cedryck Vaquette
- School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
| | - Janet Denbeigh
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sanjeev Kakar
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Simon M. Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Andre J. van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
27
|
Xue F, Zhao Z, Gu Y, Han J, Ye K, Zhang Y. 7,8-Dihydroxyflavone modulates bone formation and resorption and ameliorates ovariectomy-induced osteoporosis. eLife 2021; 10:e64872. [PMID: 34227467 PMCID: PMC8285109 DOI: 10.7554/elife.64872] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
Imbalances in bone formation and resorption cause osteoporosis. Mounting evidence supports that brain-derived neurotrophic factor (BDNF) implicates in this process. 7,8-Dihydroxyflavone (7,8-DHF), a plant-derived small molecular TrkB agonist, mimics the functions of BDNF. We show that both BDNF and 7,8-DHF promoted the proliferation, osteogenic differentiation, and mineralization of MC3T3-E1 cells. These effects might be attributed to the activation of the Wnt/β-catenin signaling pathway as the expression of cyclin D1, phosphorylated-glycogen synthase kinase-3β (p-GSK3β), β-catenin, Runx2, Osterix, and osteoprotegerin (OPG) was all significantly up-regulated. Knockdown of β-catenin restrained the up-regulation of Runx2 and Osterix stimulated by 7,8-DHF. In particular, blocking TrkB by its specific inhibitor K252a suppressed 7,8-DHF-induced osteoblastic proliferation, differentiation, and expression of osteoblastogenic genes. Moreover, BDNF and 7,8-DHF repressed osteoclastic differentiation of RAW264.7 cells. The transcription factor c-fos and osteoclastic genes such as tartrate-resistant acid phosphatase (TRAP), matrix metalloprotein-9 (MMP-9), Adamts5 were inhibited by 7,8-DHF. More importantly, 7,8-DHF attenuated bone loss, improved trabecular microarchitecture, tibial biomechanical properties, and bone biochemical indexes in an ovariectomy (OVX) rat model. The current work highlights the dual regulatory effects that 7,8-DHF exerts on bone remodeling.
Collapse
Affiliation(s)
- Fan Xue
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing; Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang UniversityHangzhouChina
| | - Zhenlei Zhao
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing; Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang UniversityHangzhouChina
| | - Yanpei Gu
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing; Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang UniversityHangzhouChina
| | - Jianxin Han
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing; Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang UniversityHangzhouChina
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of MedicineAtlantaUnited States
| | - Ying Zhang
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing; Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang UniversityHangzhouChina
| |
Collapse
|
28
|
Marupanthorn K, Tantrawatpan C, Kheolamai P, Tantikanlayaporn D, Manochantr S. MicroRNA treatment modulates osteogenic differentiation potential of mesenchymal stem cells derived from human chorion and placenta. Sci Rep 2021; 11:7670. [PMID: 33828198 PMCID: PMC8027176 DOI: 10.1038/s41598-021-87298-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/25/2021] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are important in regenerative medicine because of their potential for multi-differentiation. Bone marrow, chorion and placenta have all been suggested as potential sources for clinical application. However, the osteogenic differentiation potential of MSCs derived from chorion or placenta is not very efficient. Bone morphogenetic protein-2 (BMP-2) plays an important role in bone development. Its effect on osteogenic augmentation has been addressed in several studies. Recent studies have also shown a relationship between miRNAs and osteogenesis. We hypothesized that miRNAs targeted to Runt-related transcription factor 2 (Runx-2), a major transcription factor of osteogenesis, are responsible for regulating the differentiation of MSCs into osteoblasts. This study examines the effect of BMP-2 on the osteogenic differentiation of MSCs isolated from chorion and placenta in comparison to bone marrow-derived MSCs and investigates the role of miRNAs in the osteogenic differentiation of MSCs from these sources. MSCs were isolated from human bone marrow, chorion and placenta. The osteogenic differentiation potential after BMP-2 treatment was examined using ALP staining, ALP activity assay, and osteogenic gene expression. Candidate miRNAs were selected and their expression levels during osteoblastic differentiation were examined using real-time RT-PCR. The role of these miRNAs in osteogenesis was investigated by transfection with specific miRNA inhibitors. The level of osteogenic differentiation was monitored after anti-miRNA treatment. MSCs isolated from chorion and placenta exhibited self-renewal capacity and multi-lineage differentiation potential similar to MSCs isolated from bone marrow. BMP-2 treated MSCs showed higher ALP levels and osteogenic gene expression compared to untreated MSCs. All investigated miRNAs (miR-31, miR-106a and miR148) were consistently downregulated during the process of osteogenic differentiation. After treatment with miRNA inhibitors, ALP activity and osteogenic gene expression increased over the time of osteogenic differentiation. BMP-2 has a positive effect on osteogenic differentiation of chorion- and placenta-derived MSCs. The inhibition of specific miRNAs enhanced the osteogenic differentiation capacity of various MSCs in culture and this strategy might be used to promote bone regeneration. However, further in vivo experiments are required to assess the validity of this approach.
Collapse
Affiliation(s)
- Kulisara Marupanthorn
- Department of Agricultural Technology and Development, Faculty of Agricultural Technology, Chiangmai Rajabhat University, Chiangmai, 50330, Thailand
| | - Chairat Tantrawatpan
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.,Center of Excellence in Stem Cell Research, Thammasat University, Pathumthani, 12120, Thailand
| | - Pakpoom Kheolamai
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.,Center of Excellence in Stem Cell Research, Thammasat University, Pathumthani, 12120, Thailand
| | - Duangrat Tantikanlayaporn
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.,Center of Excellence in Stem Cell Research, Thammasat University, Pathumthani, 12120, Thailand
| | - Sirikul Manochantr
- Division of Cell Biology, Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand. .,Center of Excellence in Stem Cell Research, Thammasat University, Pathumthani, 12120, Thailand.
| |
Collapse
|
29
|
A Novel Long Noncoding RNA, Lnc-OAD, Is Required for Bone Morphogenetic Protein 2- (BMP-2-) Induced Osteoblast Differentiation. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6697749. [PMID: 33816629 PMCID: PMC7987440 DOI: 10.1155/2021/6697749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/08/2020] [Accepted: 03/03/2021] [Indexed: 01/16/2023]
Abstract
Long noncoding RNAs (lncRNAs) play very important roles in cell differentiation. Our recent study has demonstrated that a novel lncRNA named lnc-OAD modulated 3T3-L1 adipocyte differentiation. In the present study, we examined the roles of lnc-OAD in bone morphogenetic protein 2- (BMP-2-) induced osteoblast differentiation. Lnc-OAD expression was increased during BMP-2-induced osteoblast differentiation in C3H10T1/2 mesenchymal stem cells and MC3T3-E1 preosteoblast cells. Knockdown of lnc-OAD expression by specific siRNA remarkably decreased early osteoblast differentiation. In addition, stable knockdown of lnc-OAD by lentivirus vector also significantly inhibited late osteoblast differentiation and matrix mineralization in vitro. Conversely, stably overexpressed lnc-OAD with lentiviral vector accelerated osteoblast differentiation. Mechanistically, knockdown of lnc-OAD reduced significantly the phosphorylation of AKT and the expression of Osterix induced by BMP-2, while overexpression of lnc-OAD enhanced the phosphorylation of AKT and the expression of Osterix. Taken together, our study suggests that lnc-OAD plays an important role in modulating BMP-2-induced osteoblast differentiation via, at least in part, regulating the AKT-Osterix signaling axis.
Collapse
|
30
|
Multiple Myeloma Bone Disease: Implication of MicroRNAs in Its Molecular Background. Int J Mol Sci 2021; 22:ijms22052375. [PMID: 33673480 PMCID: PMC7956742 DOI: 10.3390/ijms22052375] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 12/16/2022] Open
Abstract
Multiple myeloma (MM) is a common hematological malignancy arising from terminally differentiated plasma cells. In the majority of cases, symptomatic disease is characterized by the presence of bone disease. Multiple myeloma bone disease (MMBD) is a result of an imbalance in the bone-remodeling process that leads to increased osteoclast activity and decreased osteoblast activity. The molecular background of MMBD appears intriguingly complex, as several signaling pathways and cell-to-cell interactions are implicated in the pathophysiology of MMBD. MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate the expression of their target mRNAs. Numerous miRNAs have been witnessed to be involved in cancer and hematological malignancies and their role has been characterized either as oncogenic or oncosuppressive. Recently, scientific research turned towards miRNAs as regulators of MMBD. Scientific data support that miRNAs finely regulate the majority of the signaling pathways implicated in MMBD. In this review, we provide concise information regarding the molecular pathways with a significant role in MMBD and the miRNAs implicated in their regulation. Moreover, we discuss their utility as molecular biomarkers and highlight the putative usage of miRNAs as novel molecular targets for targeted therapy in MMBD.
Collapse
|
31
|
Jeong MJ, Lim DS, Kim SO, Park C, Choi YH, Jeong SJ. Effect of rosmarinic acid on differentiation and mineralization of MC3T3-E1 osteoblastic cells on titanium surface. Anim Cells Syst (Seoul) 2021; 25:46-55. [PMID: 33717416 PMCID: PMC7935130 DOI: 10.1080/19768354.2021.1886987] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Titanium (Ti) is a widely used biomaterial for dental implants because of its outstanding biocompatibility for hard tissues. Osseointegration, the interaction between implanted biomaterials and living cells in bone, is essential for successful implantation. Rosmarinic acid (RA) is a plant-derived phytochemical with low toxicity and side effects and has various effects that can be applied as a therapeutic substance. The MC3T3-E1 osteoblastic cells on the Ti surface in medium with or without 14 μg/ml RA were used to test RA effects on osteoblast differentiation, cell viability and mineralization during differentiation. RA treatment increased osteoblast differentiation, cell viability and mineralization in MC3T3-E1 osteoblastic cells on Ti surface during differentiation, upregulating Runx-2 and OPG, but downregulating RANKL. This study suggest that RA should be applied as an effective functional and therapeutic substance to enhance osseointegration of osteoblast cells by increasing differentiation, mineralization, and bone formation through the RANKL/RANK/OPG pathway during the differentiation in MC3T3-E1 osteoblastic cells on the Ti surface.
Collapse
Affiliation(s)
- Moon-Jin Jeong
- Department of Oral Histology and Developmental Biology, School of Dentistry, Chosun University, Gwangju, Korea
| | - Do-Seon Lim
- Department of Dental Hygiene, Graduate School of Public Health Science, Eulji University, Seongnam, Korea
| | - Sung Ok Kim
- Department of Food Science and Biotechnology, College of Engineering, Kyungsung University, Busan, Korea
| | - Cheol Park
- College of Liberal Studies, Division of Basic Sciences, Dong-eui University, Busan, Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan, Korea
| | - Soon-Jeong Jeong
- Department of Dental Hygiene, College of Health Science, Youngsan University, Yangsan, Korea.,Institute of Basic Science for Well-Aging, Youngsan University, Yangsan, Korea
| |
Collapse
|
32
|
Gu X, Sun X, Sun Y, Wang J, Liu Y, Yu K, Wang Y, Zhou Y. Bioinspired Modifications of PEEK Implants for Bone Tissue Engineering. Front Bioeng Biotechnol 2021; 8:631616. [PMID: 33511108 PMCID: PMC7835420 DOI: 10.3389/fbioe.2020.631616] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022] Open
Abstract
In recent years, polyetheretherketone (PEEK) has been increasingly employed as an implant material in clinical applications. Although PEEK is biocompatible, chemically stable, and radiolucent and has an elastic modulus similar to that of natural bone, it suffers from poor integration with surrounding bone tissue after implantation. To improve the bioactivity of PEEK, numerous strategies for functionalizing the PEEK surface and changing the PEEK structure have been proposed. Inspired by the components, structure, and function of bone tissue, this review discusses strategies to enhance the biocompatibility of PEEK implants and provides direction for fabricating multifunctional implants in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
33
|
Gromolak S, Krawczenko A, Antończyk A, Buczak K, Kiełbowicz Z, Klimczak A. Biological Characteristics and Osteogenic Differentiation of Ovine Bone Marrow Derived Mesenchymal Stem Cells Stimulated with FGF-2 and BMP-2. Int J Mol Sci 2020; 21:E9726. [PMID: 33419255 PMCID: PMC7766718 DOI: 10.3390/ijms21249726] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cell-based therapies using mesenchymal stem cells (MSCs) are a promising tool in bone tissue engineering. Bone regeneration with MSCs involves a series of molecular processes leading to the activation of the osteoinductive cascade supported by bioactive factors, including fibroblast growth factor-2 (FGF-2) and bone morphogenetic protein-2 (BMP-2). In this study, we examined the biological characteristics and osteogenic differentiation potential of sheep bone marrow MSCs (BM-MSCs) treated with 20 ng/mL of FGF-2 and 100 ng/mL BMP-2 in vitro. The biological properties of osteogenic-induced BM-MSCs were investigated by assessing their morphology, proliferation, phenotype, and cytokine secretory profile. The osteogenic differentiation was characterized by Alizarin Red S staining, immunofluorescent staining of osteocalcin and collagen type I, and expression levels of genetic markers of osteogenesis. The results demonstrated that BM-MSCs treated with FGF-2 and BMP-2 maintained their primary MSC properties and improved their osteogenic differentiation capacity, as confirmed by increased expression of osteocalcin and collagen type I and upregulation of osteogenic-related gene markers BMP-2, Runx2, osterix, collagen type I, osteocalcin, and osteopontin. Furthermore, sheep BM-MSCs produced a variety of bioactive factors involved in osteogenesis, and supplementation of the culture medium with FGF-2 and BMP-2 affected the secretome profile of the cells. The results suggest that sheep osteogenic-induced BM-MSCs may be used as a cellular therapy to study bone repair in the preclinical large animal model.
Collapse
Affiliation(s)
- Sandra Gromolak
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (S.G.); (A.K.)
| | - Agnieszka Krawczenko
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (S.G.); (A.K.)
| | - Agnieszka Antończyk
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, pl. Grunwaldzki 51, 50-366 Wroclaw, Poland; (A.A.); (K.B.); (Z.K.)
| | - Krzysztof Buczak
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, pl. Grunwaldzki 51, 50-366 Wroclaw, Poland; (A.A.); (K.B.); (Z.K.)
| | - Zdzisław Kiełbowicz
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, pl. Grunwaldzki 51, 50-366 Wroclaw, Poland; (A.A.); (K.B.); (Z.K.)
| | - Aleksandra Klimczak
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (S.G.); (A.K.)
| |
Collapse
|
34
|
Safety and Efficacy Results of BonoFill First-in-Human, Phase I/IIa Clinical Trial for the Maxillofacial Indication of Sinus Augmentation and Mandibular Bone Void Filling. J Oral Maxillofac Surg 2020; 79:787-798.e2. [PMID: 33434518 DOI: 10.1016/j.joms.2020.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 11/21/2022]
Abstract
PURPOSE The gold standard for bone regeneration of bone deficiencies is still an autologous bone graft, which has considerable disadvantages; namely, the need for a second major surgery and the limited volume of bone available for harvesting. BonoFill (BF) is a novel, tissue-engineered, bone graft with intrinsic osteoinductive, osteoconductive, and osteogenic properties, consisting of the patient's own adipose tissue-derived mesenchymal stem cells, attached to hydroxyapatite particles. Here, we present the safety and efficacy results of BF first-in-human clinical study for maxillofacial bone tissue regeneration. MATERIALS AND METHODS Eleven eligible male and female subjects, aged 49-65 years, were enrolled into the clinical study in 2 clinical indications: Bone augmentation and bone void grafting in the jaws. Clinical follow-up was performed throughout a period of 6 months after BF treatment and included clinical examination, blood tests, CT scans, and biopsies collected from the transplantation site to assess chronic bone infection, changes in complete blood count, and adequate bone augmentation for implant placement. RESULTS The study results demonstrated that BF promoted adequate bone tissue regeneration without complications. Per our evaluation, there were no incidents of chronic bone infection, or significant changes in complete blood count, and the patients reported overall good health for the duration of the study. At trial end, in the sinus augmentation indication, the BF treated sites residual bone was augmented at an average of 6.36 mm (Δ new bone, n = 10) and the total bone height at the treated area was on average 11.44 mm (n = 10). In the indication of filling of bone voids, the patient's average residual bone height of 2.91 mm was 15.76 mm (n = 1) at trial end. CONCLUSIONS BF treatment was shown to be safe and resulted in newly generated bone, which provided adequate bone height for placement of dental implants. Thus, BF is a promising novel autologous bone graft for bone tissue repair.
Collapse
|
35
|
Ghavimi MA, Bani Shahabadi A, Jarolmasjed S, Memar MY, Maleki Dizaj S, Sharifi S. Nanofibrous asymmetric collagen/curcumin membrane containing aspirin-loaded PLGA nanoparticles for guided bone regeneration. Sci Rep 2020; 10:18200. [PMID: 33097790 PMCID: PMC7584591 DOI: 10.1038/s41598-020-75454-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 10/15/2020] [Indexed: 01/08/2023] Open
Abstract
The goal of the current study was to develop an asymmetric guided bone regeneration (GBR) membrane benefiting from curcumin and aspirin. The membrane was prepared using electrospinning technique and then was physic-chemically characterized by the conventional methods. The release profile of aspirin from the prepared membrane was also measured by ultraviolet spectrophotometry. Also, the antibacterial activities of the membrane was evaluated. We also assessed the in vitro effects of the prepared membrane on the biocompatibility and osteogenic differentiation of dental pulp stem cells (DPSCs), and evaluated in vivo bone regeneration using the prepared membrane in the defects created in both sides of the dog’s jaw by histology. The results from the characterization specified that the membrane was successfully prepared with monodispersed nanosized fibers, uniform network shaped morphology, negative surface charge and sustained release platform for aspirin. The membrane also showed antimicrobial effects against all tested bacteria. The presence of curcumin and aspirin in the asymmetric membrane enhanced osteogenic potential at both transcriptional and translational levels. The results of the animal test showed that the test area was completely filled with new bone after just 28 days, while the commercial membrane area remained empty. There was also a soft tissue layer above the new bone area in the test side. We suggested that the prepared membrane in this work could be used as a GBR membrane to keep soft tissue from occupying bone defects in GBR surgeries. Besides, the surgeries can be benefited from antibacterial activities and bone healing effects of this novel GBR membrane while, simultaneously, promoting bone regeneration.
Collapse
Affiliation(s)
- Mohammad Ali Ghavimi
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Bani Shahabadi
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyedhosein Jarolmasjed
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
36
|
Chen Y, Chen J, Chen J, Yu H, Zheng Y, Zhao J, Zhu J. Recent advances in seafood bioactive peptides and their potential for managing osteoporosis. Crit Rev Food Sci Nutr 2020; 62:1187-1203. [PMID: 33094645 DOI: 10.1080/10408398.2020.1836606] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Marine biodiversity provides a range of diverse biological resources, including seafoods that are rich in protein and a well-balanced amino acid composition. Previous studies have shown that peptides can improve bone formation and/or inhibit bone resorption, suggesting the potential for seafood bioactive peptides (SBPs) in development of food and pharmaceuticals for management of osteoporosis. In this review, we provided an up-to-date overview of the anti-osteoporosis activity of SBPs and describe their underlying molecular mechanisms. We focus on SBPs' development, broadening the scope and depth of research, as well as strengthening in vivo and clinical research. In vitro cell cultures and in vivo animal osteoporosis models have demonstrated the potential for seafood-derived SBPs, including fish, mollusks, crustaceans, seaweed and microalgae, in preventing osteoporosis. These peptides may act by activating the signaling pathways, such as BMP/Smads, MAPK, OPG/RANKL/RANK, and NF-κB, which are associated with modulation bone health.
Collapse
Affiliation(s)
- Yixuan Chen
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou, China
| | - Jianchu Chen
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou, China.,Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou, China
| | - Juan Chen
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou, China
| | - Huilin Yu
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou, China
| | - Yangfan Zheng
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou, China
| | - Jiawen Zhao
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou, China
| | - Jiajin Zhu
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou, China.,Ningbo Research Institute, Zhejiang University, Ningbo, China
| |
Collapse
|
37
|
Takanche JS, Kim JE, Kim JS, Yi HK. Guided bone regeneration with a gelatin layer and adenoviral delivery of c-myb enhances bone healing in rat tibia. Regen Med 2020; 15:1877-1890. [PMID: 32893751 DOI: 10.2217/rme-2019-0054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Bone healing becomes problematic during certain states, such as trauma. This study verifies whether the application of c-myb with gelatin promotes bone healing during bone injuries. Materials & methods: A biodegradable membrane was modified with adenoviral vector c-myb (Ad/c-myb) and gelatin and applied in the bone injury site of rat tibia. Results: c-myb enhanced osteogenic differentiation and mineralization in bone marrow stromal cells after induction with osteogenic media. In vivo examination of rat tibia after application of the biodegradable membrane with Ad/c-myb and a gelatin layer demonstrated increased bone volume, bone mineral density, new bone formation and osteogenic molecules, compared with Ad/LacZ. Conclusion: c-myb has the potential to assist bone healing and may be applicable to the treatment of bone during injury.
Collapse
Affiliation(s)
- Jyoti Shrestha Takanche
- Departments of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju, Korea
| | - Ji-Eun Kim
- Departments of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju, Korea
| | - Jeong-Seok Kim
- Departments of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju, Korea
| | - Ho-Keun Yi
- Departments of Oral Biochemistry, Institute of Oral Bioscience, School of Dentistry, Jeonbuk National University, Jeonju, Korea
| |
Collapse
|
38
|
Local Wnt3a treatment restores bone regeneration in large osseous defects after surgical debridement of osteomyelitis. J Mol Med (Berl) 2020; 98:897-906. [PMID: 32424558 PMCID: PMC8526481 DOI: 10.1007/s00109-020-01924-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/13/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022]
Abstract
Impaired bone homeostasis caused by osteomyelitis provokes serious variations in the bone remodeling process, thereby involving multiple inflammatory cytokines to activate bone healing. We have previously established a mouse model for post-traumatic osteomyelitis and studied bone regeneration after sufficient debridement. Moreover, we could further characterize the postinfectious inflammatory state of bony defects after debridement with elevated osteoclasts and decreased bone formation despite the absence of bacteria. In this study, we investigated the positive effects of Wnt-pathway modulation on bone regeneration in our previous established mouse model. This was achieved by local application of Wnt3a, a recombinant activator of the canonical Wnt-pathway. Application of Wnt3a could enhance new bone formation, which was verified by histological and μ-CT analysis. Moreover, histology and western blots revealed enhanced osteoblastogenesis and downregulated osteoclasts in a RANKL-dependent manner. Further analysis of Wnt-pathway showed downregulation after bone infections were reconstituted by application of Wnt3a. Interestingly, Wnt-inhibitory proteins Dickkopf 1 (DKK1), sclerostin, and secreted frizzled protein 1 (sFRP1) were upregulated simultaneously to Wnt-pathway activation, indicating a negative feedback for active form of Beta-catenin. In this study, we could demonstrate enhanced bone formation in defects caused by post-traumatic osteomyelitis after Wnt3a application. KEY MESSAGES: Osteomyelitis decreases bone regeneration Wnt3a restores bone healing after infection Canonical Wnt-pathway activation with negative feedback.
Collapse
|
39
|
Zhu S, Bennett S, Kuek V, Xiang C, Xu H, Rosen V, Xu J. Endothelial cells produce angiocrine factors to regulate bone and cartilage via versatile mechanisms. Am J Cancer Res 2020; 10:5957-5965. [PMID: 32483430 PMCID: PMC7255007 DOI: 10.7150/thno.45422] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Blood vessels are conduits distributed throughout the body, supporting tissue growth and homeostasis by the transport of cells, oxygen and nutrients. Endothelial cells (ECs) form the linings of the blood vessels, and together with pericytes, are essential for organ development and tissue homeostasis through producing paracrine signalling molecules, called angiocrine factors. In the skeletal system, ECs - derived angiocrine factors, combined with bone cells-released angiogenic factors, orchestrate intercellular crosstalk of the bone microenvironment, and the coupling of angiogenesis-to-osteogenesis. Whilst the involvement of angiogenic factors and the blood vessels of the skeleton is relatively well established, the impact of ECs -derived angiocrine factors on bone and cartilage homeostasis is gradually emerging. In this review, we survey ECs - derived angiocrine factors, which are released by endothelial cells of the local microenvironment and by distal organs, and act specifically as regulators of skeletal growth and homeostasis. These may potentially include angiocrine factors with osteogenic property, such as Hedgehog, Notch, WNT, bone morphogenetic protein (BMP), fibroblast growth factor (FGF), insulin-like growth factor (IGF), and platelet-derived growth factor (PDGF). Understanding the versatile mechanisms by which ECs-derived angiocrine factors orchestrate bone and cartilage homeostasis, and pathogenesis, is an important step towards the development of therapeutic potential for skeletal diseases.
Collapse
|
40
|
Dudakovic A, Samsonraj RM, Paradise CR, Galeano-Garces C, Mol MO, Galeano-Garces D, Zan P, Galvan ML, Hevesi M, Pichurin O, Thaler R, Begun DL, Kloen P, Karperien M, Larson AN, Westendorf JJ, Cool SM, van Wijnen AJ. Inhibition of the epigenetic suppressor EZH2 primes osteogenic differentiation mediated by BMP2. J Biol Chem 2020; 295:7877-7893. [PMID: 32332097 DOI: 10.1074/jbc.ra119.011685] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/22/2020] [Indexed: 12/17/2022] Open
Abstract
Bone-stimulatory therapeutics include bone morphogenetic proteins (e.g. BMP2), parathyroid hormone, and antibody-based suppression of WNT antagonists. Inhibition of the epigenetic enzyme enhancer of zeste homolog 2 (EZH2) is both bone anabolic and osteoprotective. EZH2 inhibition stimulates key components of bone-stimulatory signaling pathways, including the BMP2 signaling cascade. Because of high costs and adverse effects associated with BMP2 use, here we investigated whether BMP2 dosing can be reduced by co-treatment with EZH2 inhibitors. Co-administration of BMP2 with the EZH2 inhibitor GSK126 enhanced differentiation of murine (MC3T3) osteoblasts, reflected by increased alkaline phosphatase activity, Alizarin Red staining, and expression of bone-related marker genes (e.g. Bglap and Phospho1). Strikingly, co-treatment with BMP2 (10 ng/ml) and GSK126 (5 μm) was synergistic and was as effective as 50 ng/ml BMP2 at inducing MC3T3 osteoblastogenesis. Similarly, the BMP2-GSK126 co-treatment stimulated osteogenic differentiation of human bone marrow-derived mesenchymal stem/stromal cells, reflected by induction of key osteogenic markers (e.g. Osterix/SP7 and IBSP). A combination of BMP2 (300 ng local) and GSK126 (5 μg local and 5 days of 50 mg/kg systemic) yielded more consistent bone healing than single treatments with either compound in a mouse calvarial critical-sized defect model according to results from μCT, histomorphometry, and surgical grading of qualitative X-rays. We conclude that EZH2 inhibition facilitates BMP2-mediated induction of osteogenic differentiation of progenitor cells and maturation of committed osteoblasts. We propose that epigenetic priming, coupled with bone anabolic agents, enhances osteogenesis and could be leveraged in therapeutic strategies to improve bone mass.
Collapse
Affiliation(s)
- Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA.,Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Christopher R Paradise
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Merel O Mol
- Department of Orthopedic Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Pengfei Zan
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA.,Department of Orthopedic Surgery, School of Medicine, Second Affiliated Hospital of Zhejiang University, Hangzhou, China.,Department of Orthopedic Surgery, School of Medicine, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - M Lizeth Galvan
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Mario Hevesi
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Oksana Pichurin
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Dana L Begun
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter Kloen
- Department of Orthopedic Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| | - A Noelle Larson
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA.,Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA .,Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
41
|
Gonçalves FC, Oliveira GJPLD, Scardueli CR, Spin-Neto R, Stavropoulos A, Marcantonio RAC. Cyclosporine A impairs bone repair in critical defects filled with different osteoconductive bone substitutes. Braz Oral Res 2020; 34:e007. [PMID: 32049108 DOI: 10.1590/1807-3107bor-2020.vol34.0007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 11/01/2019] [Indexed: 12/29/2022] Open
Abstract
The aim of this study was to assess the influence of cyclosporine administration on the repair of critical-sized calvaria defects (CSDs) in rat calvaria filled with diverse biomaterials. Sixty animals were divided into two groups: the control (CTR) group (saline solution) and the cyclosporine (CCP) group (cyclosporine, 10 mg/kg/day). These medications were administered daily by gavage, beginning 15 days before the surgical procedure and lasting until the day the animals were euthanized. A CSD (5 mm Ø) was made in the calvaria of each animal, which was allocated to one of 3 subgroups, according to the biomaterial used to fill the defect: coagulum (COA), deproteinized bovine bone (DBB), or biphasic calcium phosphate ceramics of hydroxyapatite and β-phosphate tricalcium (HA/TCP). Euthanasia of the animals was performed 15 and 60 days after the surgical procedure (n = 5 animals/period/subgroup). Bone repair (formation) assessment was performed through microtomography and histometry, while the analyses of the expression of the BMP2, Osteocalcin, and TGFβ1 proteins were performed using immunohistochemistry. The CSDs not filled with biomaterials demonstrated lower bone formation in the CCP group. At 15 days, less bone formation was observed in the CSDs filled with DBB, a smaller volume of mineralized tissue was observed in the CSDs filled with HA/TCP, and the expression levels of BMP2 and osteocalcin were lower in the CCP group compared to the CTR group. The use of cyclosporine impaired bone repair in CSD, and this effect can be partially explained by the suppression of BMP2 and osteocalcin expression.
Collapse
Affiliation(s)
- Fernanda Castanheira Gonçalves
- Universidade Estadual de São Paulo - Unesp, School of Dentistry Araraquara, Department of Diagnosis and Surgery, Araraquara, SP, Brazil
| | | | - Cassio Rocha Scardueli
- Universidade Estadual de São Paulo - Unesp, School of Dentistry Araraquara, Department of Diagnosis and Surgery, Araraquara, SP, Brazil
| | - Rubens Spin-Neto
- Aarhus University, Department of Dentistry and Oral Health, Section of Oral Radiology, Aarhus, Denmark
| | - Andreas Stavropoulos
- Malmo University, Department of Community Dentistry and Periodontology, Malmö, Sweden
| | | |
Collapse
|
42
|
Tang L, Kang Y, Sun S, Zhao T, Cao W, Fan X, Guo J, Sun L, Ta D. Inhibition of MSTN signal pathway may participate in LIPUS preventing bone loss in ovariectomized rats. J Bone Miner Metab 2020; 38:14-26. [PMID: 31414284 DOI: 10.1007/s00774-019-01029-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/06/2019] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Menopause can lead to osteoporosis, which is characterized by destruction of bone microstructure, poor mechanical properties, and prone to fracture. LIPUS can effectively promote bone formation and fracture healing. MSTN is a transforming growth factor-β family member that acts as a negative regulator of skeletal muscle growth. A MSTN deficiency also has a positive effect on bone formation. However, whether LIPUS could inhibit bone loss and promote healing of bone injury of menopause through the inhibition of the MSTN signaling pathway has not been previously investigated. We herein investigated the effects of LIPUS on bone architecture, mechanical properties, the healing of bone defects, and its potential molecular mechanisms in ovariectomized rats. MATERIALS AND METHODS The rats were randomly divided into three groups: sham ovariectomized group (Sham), ovariectomized model group (OVX), ovariectomized model with LIPUS therapy group (OVX + LIPUS). The OVX + LIPUS rats were treated with LIPUS (1.5 MHz, 30 mW/cm2) on the femur for 20 min/day that lasted for 19 days. RESULTS LIPUS effectively improved the bone microstructure, increased mechanical properties and promoted the healing of bone defects in ovariectomized rats. Moreover, LIPUS effectively decreased the MSTN content in serum and quadriceps muscle in ovariectomized rats, and inhibited the expression of MSTN downstream signaling molecules and activated the Wnt signaling pathway in the femur. CONCLUSIONS The present study shows that LIPUS improved osteoporosis and promoted bone defect healing in the ovariectomized rats may through the inhibition of the MSTN signal pathway.
Collapse
Affiliation(s)
- Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Yiting Kang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shuxin Sun
- Department of Electronic Engineering, Fudan University, Shanghai, 200433, China
| | - Tingting Zhao
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Wenxin Cao
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Jianzhong Guo
- Shaanxi Key Laboratory of Ultrasonics, Shaanxi Normal University, Xi'an, 710119, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China.
| | - Dean Ta
- Department of Electronic Engineering, Fudan University, Shanghai, 200433, China.
- Human Phenome Institute, Fudan University, Shanghai, 201203, China.
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention (MICCAI) of Shanghai, Shanghai, 200032, China.
| |
Collapse
|
43
|
Tarafder S, Brito JA, Minhas S, Effiong L, Thomopoulos S, Lee CH. In situ tissue engineering of the tendon-to-bone interface by endogenous stem/progenitor cells. Biofabrication 2019; 12:015008. [PMID: 31561236 PMCID: PMC6904927 DOI: 10.1088/1758-5090/ab48ca] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The long-term success of surgical repair of rotator cuff tears is largely dependent on restoration of a functional tendon-to-bone interface. We implemented micro-precise spatiotemporal delivery of growth factors in three-dimensional printed scaffolds for integrative regeneration of a fibrocartilaginous tendon-to-bone interface. Sustained and spatially controlled release of tenogenic, chondrogenic and osteogenic growth factors was achieved using microsphere-based delivery carriers embedded in thin membrane-like scaffolds. In vitro, the scaffolds embedded with spatiotemporal delivery of growth factors successfully guided regional differentiation of mesenchymal progenitor cells, forming multiphase tissues with tendon-like, cartilage-like and bone-like regions. In vivo, when implanted at the interface between the supraspinatus tendon and the humeral head in a rat rotator cuff repair model, these scaffolds promoted recruitment of endogenous tendon progenitor cells followed by integrative healing of tendon and bone via re-formation of strong fibrocartilaginous interfaces. Our findings demonstrate the potential of in situ tissue engineering of tendon-to-bone interfaces by endogenous progenitor cells. The in situ tissue engineering approach shows translational potential for improving outcomes after rotator cuff repair.
Collapse
Affiliation(s)
- Solaiman Tarafder
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168th Street, VC12-230, NY 10032, New York
| | - John A Brito
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168th Street, VC12-230, NY 10032, New York
| | - Sumeet Minhas
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168th Street, VC12-230, NY 10032, New York
| | - Linda Effiong
- Department of Orthopedic Surgery, Columbia University Medical Center, 650 W. 168th Street, BB14-1408, NY 10032, New York
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University Medical Center, 650 W. 168th Street, BB14-1408, NY 10032, New York
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, NY 10027, New York
| | - Chang H Lee
- Regenerative Engineering Laboratory, Columbia University Medical Center, 630 W. 168th Street, VC12-230, NY 10032, New York
| |
Collapse
|
44
|
Yanai R, Tetsuo F, Ito S, Itsumi M, Yoshizumi J, Maki T, Mori Y, Kubota Y, Kajioka S. Extracellular calcium stimulates osteogenic differentiation of human adipose-derived stem cells by enhancing bone morphogenetic protein-2 expression. Cell Calcium 2019; 83:102058. [DOI: 10.1016/j.ceca.2019.102058] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 06/19/2019] [Accepted: 07/16/2019] [Indexed: 12/17/2022]
|
45
|
The growth of the posterior cranial fossa in FGFR2-induced faciocraniosynostosis: A review. Neurochirurgie 2019; 65:221-227. [PMID: 31557489 DOI: 10.1016/j.neuchi.2019.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/25/2019] [Accepted: 09/12/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND The growth of the posterior fossa in syndromic craniostenosis was studied in many papers. However, few studies described the pathophysiological growth mechanisms in non-operated infants with fibroblast growth factor receptor (FGFR) type 2 mutation (Crouzon, Apert or Pfeiffer syndrome), although these are essential to understanding cranial vault expansion and hydrocephalus treatment in these syndromes. OBJECTIVE A review of the medical literature was performed, to understand the physiological and pathological growth mechanisms of the posterior fossa in normal infants and infants with craniostenosis related to FGFR2 mutation. DISCUSSION Of the various techniques for measuring posterior fossa volume, direct slice-by-slice contouring is the most precise and sensitive. Posterior fossa growth follows a bi-phasic pattern due to opening of the petro-occipital, occipitomastoidal and spheno-occipital sutures. Some studies reported smaller posterior fossae in syndromic craniostenosis, whereas direct contouring studies reported no difference between normal and craniostenotic patients. In Crouzon syndrome, synchondrosis fusion occurs earlier than in normal subjects, and follows a precise pattern. This premature fusion in Crouzon syndrome leads to a stenotic foramen magnum and facial retrusion.
Collapse
|
46
|
Sheyn D, Cohn-Yakubovich D, Ben-David S, De Mel S, Chan V, Hinojosa C, Wen N, Hamilton GA, Gazit D, Gazit Z. Bone-chip system to monitor osteogenic differentiation using optical imaging. MICROFLUIDICS AND NANOFLUIDICS 2019; 23:99. [PMID: 32296299 PMCID: PMC7158882 DOI: 10.1007/s10404-019-2261-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Human organoids and organ-on-chip systems to predict human responses to new therapies and for the understanding of disease mechanisms are being more commonly used in translational research. We have developed a bone-chip system to study osteogenic differentiation in vitro, coupled with optical imaging approach which provides the opportunity of monitoring cell survival, proliferation and differentiation in vitro without the need to terminate the culture. We used the mesenchymal stem cell (MSC) line over-expressing bone morphogenetic protein-2 (BMP-2), under Tet-Off system, and luciferase reporter gene under constitutive promoter. Cells were seeded on chips and supplemented with osteogenic medium. Flow of media was started 24 h later, while static cultures were performed using media reservoirs. Cells grown on the bone-chips under constant flow of media showed enhanced survival/proliferation, comparing to the cells grown in static conditions; luciferase reporter gene expression and activity, reflecting the cell survival and proliferation, was quantified using bioluminescence imaging and a significant advantage to the flow system was observed. In addition, the flow had positive effect on osteogenic differentiation, when compared with static cultures. Quantitative fluorescent imaging, performed using the osteogenic extra-cellular matrix-targeted probes, showed higher osteogenic differentiation of the cells under the flow conditions. Gene expression analysis of osteogenic markers confirmed the osteogenic differentiation of the MSC-BMP2 cells. Immunofluorescent staining performed against the Osteocalcin, Col1, and BSP markers illustrated robust osteogenic differentiation in the flow culture and lessened differentiation in the static culture. To sum, the bone-chip allows monitoring cell survival, proliferation, and osteogenic differentiation using optical imaging.
Collapse
Affiliation(s)
- Dmitriy Sheyn
- Orthopedic Stem Cell Research Lab, Cedars-Sinai Medical Center, AHSP-A8308, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Orthopaedics, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
| | - Doron Cohn-Yakubovich
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, 91120 Jerusalem, Israel
| | - Shiran Ben-David
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Skeletal Regeneration Program, Cedars-Sinai Medical Center, AHSP-8304, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Sandra De Mel
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Skeletal Regeneration Program, Cedars-Sinai Medical Center, AHSP-8304, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Virginia Chan
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Skeletal Regeneration Program, Cedars-Sinai Medical Center, AHSP-8304, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | | | | | | | - Dan Gazit
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Orthopaedics, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, 91120 Jerusalem, Israel
- Skeletal Regeneration Program, Cedars-Sinai Medical Center, AHSP-8304, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Zulma Gazit
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Orthopaedics, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles 90048, CA, USA
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, 91120 Jerusalem, Israel
- Skeletal Regeneration Program, Cedars-Sinai Medical Center, AHSP-8304, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| |
Collapse
|
47
|
Nah H, Lee D, Heo M, Lee JS, Lee SJ, Heo DN, Seong J, Lim HN, Lee YH, Moon HJ, Hwang YS, Kwon IK. Vitamin D-conjugated gold nanoparticles as functional carriers to enhancing osteogenic differentiation. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2019; 20:826-836. [PMID: 31489055 PMCID: PMC6713151 DOI: 10.1080/14686996.2019.1644193] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 05/09/2023]
Abstract
In an aging society, bone disorders such as osteopenia, osteoporosis, and degenerative arthritis cause serious public health problems. In order to solve these problems, researchers continue to develop therapeutic agents, increase the efficacy of developed therapeutic agents, and reduce side effects. Gold nanoparticles (GNPs) are widely used in tissue engineering applications as biosensors, drug delivery carriers, and bioactive materials. Their special surface property enables easy conjugation with ligands including functional groups such as thiols, phosphines, and amines. This creates an attractive advantage to GNPs for use in the bone tissue engineering field. However, GNPs alone are limited in their biological effects. In this study, we used thiol-PEG-vitamin D (SPVD) to conjugate vitamin D, an essential nutrient critical for maintaining normal skeletal homeostasis, to GNPs. To characterize vitamin D-conjugated GNPs (VGNPs), field emission transmission electron microscopy, energy dispersive X-ray spectroscopy, dynamic light scattering, and ultraviolet/visible absorption analysis were carried out. The developed VGNPs were well bound through the thiol groups between GNPs and vitamin D, and were fabricated in size of 60 nm. Moreover, to demonstrate VGNPs osteogenic differentiation effect, various assays were carried out through cell viability test, alkaline phosphatase assay, calcium deposition assay, real-time polymerase chain reaction, and immunofluorescence staining. As a result, the fabricated VGNPs were found to effectively enhance osteogenic differentiation of human adipose-derived stem cells (hADSCs) in vitro. Based on these results, VGNPs can be utilized as functional nanomaterials for bone regeneration in the tissue engineering field.
Collapse
Affiliation(s)
- Haram Nah
- Department of Dentistry, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Donghyun Lee
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Min Heo
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Jae Seo Lee
- Department of Dentistry, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Sang Jin Lee
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Dong Nyoung Heo
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Jeongmin Seong
- Department of Dental Hygiene, College of Health Science, Kangwon National University, Samcheok-si, Republic of Korea
| | - Ho-Nam Lim
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Yeon-Hee Lee
- Department of Orofacial Pain and Oral Medicine, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Ho-Jin Moon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Yu-Shik Hwang
- Department of Maxillofacial Biomedical Engineering, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
48
|
Peroxiredoxin II negatively regulates BMP2-induced osteoblast differentiation and bone formation via PP2A Cα-mediated Smad1/5/9 dephosphorylation. Exp Mol Med 2019; 51:1-11. [PMID: 31160554 PMCID: PMC6546700 DOI: 10.1038/s12276-019-0263-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/08/2019] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
Peroxiredoxin II (Prx II), an antioxidant enzyme in the Prx family, reduces oxidative stress by decreasing the intracellular ROS levels. Osteoblast differentiation is promoted by bone morphogenetic protein 2 (BMP2), which upregulates the expression of osteoblast differentiation marker genes, through Smad1/5/9 phosphorylation. We found that Prx II expression was increased by a high dose of lipopolysaccharide (LPS) but was not increased by a low dose of LPS. Prx II itself caused a decrease in the osteogenic gene expression, alkaline phosphatase (ALP) activity, and Smad1/5/9 phosphorylation induced by BMP2. In addition, BMP2-induced osteogenic gene expression and ALP activity were higher in Prx II knockout (KO) cells than they were in wild-type (WT) cells. These inhibitory effects were mediated by protein phosphatase 2A Cα (PP2A Cα), which was increased and is known to induce the dephosphorylation of Smad1/5/9. The overexpression of Prx II increased the expression of PP2A Cα, and PP2A Cα was not expressed in Prx II KO cells. Moreover, PP2A Cα reduced the level of BMP2-induced osteogenic gene expression and Smad1/5/9 phosphorylation. LPS inhibited BMP2-induced Smad1/5/9 phosphorylation and the suppressed phosphorylation was restored by the PP2A inhibitor okadaic acid (OA). Bone phenotype analyses using microcomputed tomography (μCT) revealed that the Prx II KO mice had higher levels of bone mass than the levels of the WT mice. We hypothesize that Prx II has a negative role in osteoblast differentiation through the PP2A-dependent dephosphorylation of Smad1/5/9. An antioxidant enzyme actively works to reduce bone synthesis under oxidative stress conditions in order to protect bone cells from damage and cell death. Bone is generated by cells called osteoblasts, which differentiate from stem cells. In osteoporosis and diabetes, excessive reactive oxygen species (ROS) within cells can disrupt osteoblast differentiation. South Korean researchers led by Eun-jung Kim at Kyungpook National University, Daegu, and Won-Gu Jang at Daegu University, Gyeongbuk, have shown that an antioxidant enzyme, peroxiredoxin II (PrxII), helps regulate bone formation under oxidative stress. The team generated PrxII-deficient mice and compared them with healthy normal mice. Under oxidative stress conditions, the mice had higher bone mass and higher expression of genes related to bone formation than the normal mice. PrxII limits osteoblast differentiation during elevated ROS by influencing associated protein activity and signalling pathways.
Collapse
|
49
|
Kim D, Thangavelu M, Cheolui S, Kim HS, Choi MJ, Song JE, Khang G. Effect of different concentration of demineralized bone powder with gellan gum porous scaffold for the application of bone tissue regeneration. Int J Biol Macromol 2019; 134:749-758. [PMID: 31054303 DOI: 10.1016/j.ijbiomac.2019.04.184] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 12/18/2022]
Abstract
The prevalence of bone-related diseases has increased, the population growth as a result of the aging phenomenon requires more effective treatments for regeneration of bone defect. Although an autogenous bone graft was used in traditional operation method, they are very inefficient in current bone defect surgery and very difficult to gather the required amount of bone for operation. It is becoming a gradually growing disease, hence there is a need for developing a new method for preparing biomimetic scaffolds. DBP (demineralized bone powder), a potent bone regeneration material, has a trace amount of ions and bone mineral component. Especially, GD (Gallus gallus var domesticus) DBP has a unique property, which has melanin, for strengthening bones, increasing ALP activity and bone mineralization, compared to other available biomaterials. For that reason, GD DBP was combined with GG (gellan gum). The material was characterized in vitro and in vivo rat model. The first priority in this work was given to assessing the attachment and proliferation rates of BMSCs following the in vivo experiment in rats. The results of 1% sample showed better osteogenic effects that can be used in clinical application after studying in larger animals for better bone regeneration and tissue engineering.
Collapse
Affiliation(s)
- David Kim
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology, Polymer Materials Fusion Research Center, Chonbuk National University, Deokjin-gu, Jeonju 561-756, Republic of Korea
| | - Muthukumar Thangavelu
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology, Polymer Materials Fusion Research Center, Chonbuk National University, Deokjin-gu, Jeonju 561-756, Republic of Korea
| | - Song Cheolui
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology, Polymer Materials Fusion Research Center, Chonbuk National University, Deokjin-gu, Jeonju 561-756, Republic of Korea
| | - Han Sol Kim
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology, Polymer Materials Fusion Research Center, Chonbuk National University, Deokjin-gu, Jeonju 561-756, Republic of Korea
| | - Min Joung Choi
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology, Polymer Materials Fusion Research Center, Chonbuk National University, Deokjin-gu, Jeonju 561-756, Republic of Korea
| | - Jeong Eun Song
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology, Polymer Materials Fusion Research Center, Chonbuk National University, Deokjin-gu, Jeonju 561-756, Republic of Korea
| | - Gilson Khang
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology, Polymer Materials Fusion Research Center, Chonbuk National University, Deokjin-gu, Jeonju 561-756, Republic of Korea.
| |
Collapse
|
50
|
MacKenzie RK, Sankar PR, Bendall AJ. Dlx5 and Dlx6 can antagonize cell division at the G 1/S checkpoint. BMC Mol Cell Biol 2019; 20:8. [PMID: 31041891 PMCID: PMC6460778 DOI: 10.1186/s12860-019-0191-6] [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: 09/13/2018] [Accepted: 04/02/2019] [Indexed: 11/17/2022] Open
Abstract
Background Dlx5 and Dlx6 stimulate differentiation of diverse progenitors during embryonic development. Their actions as pro-differentiation transcription factors includes the up-regulation of differentiation markers but the extent to which differentiation may also be stimulated by regulation of the cell cycle has not been addressed. Results We document that expression of Dlx5 and Dlx6 antagonizes cell proliferation in a variety of cell types without inducing apoptosis or promoting cell cycle exit. Rather, a variety of evidence indicates that elevated Dlx5 and Dlx6 expression reduces the proportion of cells in S phase and affects the length of the cell cycle. Conclusions Antagonism of S-phase entry by Dlx5 and Dlx6 proteins likely represents a lineage-independent function to effect Dlx-mediated differentiation in multiple progenitor cell types.
Collapse
Affiliation(s)
- Rachel K MacKenzie
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd East, Guelph, Ontario, N1G 2W1, Canada
| | - Parvathy Ravi Sankar
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd East, Guelph, Ontario, N1G 2W1, Canada
| | - Andrew J Bendall
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd East, Guelph, Ontario, N1G 2W1, Canada.
| |
Collapse
|