1
|
Yan J, Gu X, Gao X, Shao Y, Ji M. USP36 regulates the proliferation, survival, and differentiation of hFOB1.19 osteoblast. J Orthop Surg Res 2024; 19:483. [PMID: 39152465 PMCID: PMC11330066 DOI: 10.1186/s13018-024-04893-8] [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: 05/21/2024] [Accepted: 07/02/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Effective bone formation relies on osteoblast differentiation, a process subject to intricate post-translational regulation. Ubiquitin-specific proteases (USPs) repress protein degradation mediated by the ubiquitin-proteasome pathway. Several USPs have been documented to regulate osteoblast differentiation, but whether other USPs are involved in this process remains elusive. METHODS In this study, we conducted a comparative analysis of 48 USPs in differentiated and undifferentiated hFOB1.19 osteoblasts, identifying significantly upregulated USPs. Subsequently, we generated USP knockdown hFOB1.19 cells and evaluated their osteogenic differentiation using Alizarin red staining. We also assessed cell viability, cell cycle progression, and apoptosis through MTT, 7-aminoactinomycin D staining, and Annexin V/PI staining assays, respectively. Quantitative PCR and Western blotting were employed to measure the expression levels of osteogenic differentiation markers. Additionally, we investigated the interaction between the USP and its target protein using co-immunoprecipitation (co-IP). Furthermore, we depleted the USP in hFOB1.19 cells to examine its effect on the ubiquitination and stability of the target protein using immunoprecipitation (IP) and Western blotting. Finally, we overexpressed the target protein in USP-deficient hFOB1.19 cells and evaluated its impact on their osteogenic differentiation using Alizarin red staining. RESULTS USP36 is the most markedly upregulated USP in differentiated hFOB1.19 osteoblasts. Knockdown of USP36 leads to reduced viability, cell cycle arrest, heightened apoptosis, and impaired osteogenic differentiation in hFOB1.19 cells. USP36 interacts with WD repeat-containing protein 5 (WDR5), and the knockdown of USP36 causes an increased level of WDR5 ubiquitination and accelerated degradation of WDR5. Excessive WDR5 improved the impaired osteogenic differentiation of USP36-deficient hFOB1.19 cells. CONCLUSIONS These observations suggested that USP36 may function as a key regulator of osteoblast differentiation, and its regulatory mechanism may be related to the stabilization of WDR5.
Collapse
Affiliation(s)
- Junfa Yan
- Department of Orthopaedics, Xiamen Humanity Hospital, Xiamen, 361006, Fujian, China
| | - Xiufei Gu
- Department of ICU, Xiang'an Hospital of Xiamen University, Xiamen, 361100, Fujian, China
| | - Xilin Gao
- Department of Orthopaedics, Xiamen Humanity Hospital, Xiamen, 361006, Fujian, China
| | - Yan Shao
- Department of Internal Medicine, Alashan High-tech Industrial Development Zone General Hospital, Alashan, 750300, Inner Mongolia Autonomous Region, China
| | - Minghua Ji
- Department of Orthopaedics, Xiamen Humanity Hospital, Xiamen, 361006, Fujian, China.
| |
Collapse
|
2
|
Nogueira-Júnior V, Sousa FRN, da S M Rebouças C, Braz HLB, Dos S Morais MLG, Goes P, de C Brito GA, Jorge RJB, Barbosa FG, Mafezoli J, Silva-Filho CJA, de O Capistrano AL, Bezerra MM, de C Leitão RF. Exploring the osteogenic potential of semisynthetic triterpenes from Combretum leprosum: An in vitro and in silico study. In Vitro Cell Dev Biol Anim 2024:10.1007/s11626-024-00928-w. [PMID: 38992216 DOI: 10.1007/s11626-024-00928-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 05/13/2024] [Indexed: 07/13/2024]
Abstract
Combretum leprosum Mart. is a plant of the Combretaceae family, widely distributed in the Northeast region of Brazil, popularly used as an anti-inflammatory agent, and rich in triterpenes. This study evaluated in vitro and in silico potential osteogenic of two semisynthetic triterpenes (CL-P2 and CL-P2A) obtained from the pentacyclic triterpene 3β,6β,16β-trihydroxylup-20(29)-ene (CL-1) isolated from C. leprosum. Assays were carried out in cultured murine osteoblasts (OFCOL II), first investigating the possible toxicity of the compounds on these cells through viability assays (MTT). Cell proliferation and activation were investigated by immunohistochemical evaluation of Ki-67, bone alkaline phosphatase (ALP) activity, and mineralization test by Von Kossa. Molecular docking analysis was performed to predict the binding affinity of CL-P2 and CL-P2A to target proteins involved in the regulation of osteogenesis, including: bone morphogenetic protein 2 (BMP-2), proteins related to Wingless-related integration (WNT) pathway (Low-density lipoprotein receptor-related protein 6-LRP6 and sclerostin-SOST), and receptor activator of nuclear factor (NF)-kB-ligand (RANK-L). Next, Western Blot and immunofluorescence investigated BMP-2, WNT, RANK-L, and OPG protein expressions in cultured murine osteoblasts (OFCOL II). None of the CL-P2 and CL-P2A concentrations were toxic to osteoblasts. Increased cell proliferation, ALP activity, and bone mineralization were observed. Molecular docking assays demonstrated interactions with BMP-2, LRP6, SOST, and RANK-L/OPG. There was observed increased expression of BMP-2, WNT, and RANK-L/OPG proteins. These results suggest, for the first time, the osteogenic potential of CL-P2 and CL-P2A.
Collapse
Affiliation(s)
- Valdo Nogueira-Júnior
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Fátima Regina N Sousa
- Department of Morphology, Medical School, Federal University of Piaui, Picos, Piauí, Brazil
| | - Conceição da S M Rebouças
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Helyson L B Braz
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Maria Luana G Dos S Morais
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Paula Goes
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Gerly Anne de C Brito
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Roberta Jeane B Jorge
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Francisco Geraldo Barbosa
- Department of Organic and Inorganic Chemistry, Science Centre, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Jair Mafezoli
- Department of Organic and Inorganic Chemistry, Science Centre, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Carlos José A Silva-Filho
- Department of Organic and Inorganic Chemistry, Science Centre, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - André Luiz de O Capistrano
- Department of Organic and Inorganic Chemistry, Science Centre, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Mirna M Bezerra
- Postgraduate Program in Health Sciences, School of Medicine, Federal University of Ceará, Sobral, Ceará, Brazil.
| | - Renata F de C Leitão
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
3
|
Karaca MA, Kancagi DD, Ozbek U, Ovali E, Gok O. Betulin Stimulates Osteogenic Differentiation of Human Osteoblasts-Loaded Alginate-Gelatin Microbeads. Bioengineering (Basel) 2024; 11:553. [PMID: 38927789 PMCID: PMC11201098 DOI: 10.3390/bioengineering11060553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/06/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Osteoporosis, a terminal illness, has emerged as a global public health problem in recent years. The long-term use of bone anabolic drugs to treat osteoporosis causes multi-morbidity in elderly patients. Alternative therapies, such as allogenic and autogenic tissue grafts, face important issues, such as a limited source of allogenic grafts and tissue rejection in autogenic grafts. However, stem cell therapy has been shown to increase bone regeneration and decrease osteoporotic bone formation. Stem cell therapy combined with betulin (BET) supplementation might be adequate for bone remodeling and new bone tissue generation. In this study, the effect of BET on the viability and osteogenic differentiation of hFOB 1.19 cells was investigated. The cells were encapsulated in alginate-gelatin (AlGel) microbeads. In vitro tests were conducted during the 12 d of incubation. While BET showed cytotoxic activity (>1 µM) toward non-encapsulated hFOB 1.19 cells, encapsulated cells retained their functionality for up to 12 days, even at 5 µM BET. Moreover, the expression of osteogenic markers indicates an enhanced osteo-inductive effect of betulin on encapsulated hFOB 1.19, compared to the non-encapsulated cell culture. The 3D micro-environment of the AlGel microcapsules successfully protects the hFOB 1.19 cells against BET cytotoxicity, allowing BET to improve the mineralization and differentiation of osteoblast cells.
Collapse
Affiliation(s)
- Mehmet Ali Karaca
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | - Derya Dilek Kancagi
- Acibadem Labcell Cellular Therapy Laboratory, 34752 Istanbul, Turkey; (D.D.K.); (E.O.)
| | - Ugur Ozbek
- Medical Genetics Department, School of Medicine, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | - Ercument Ovali
- Acibadem Labcell Cellular Therapy Laboratory, 34752 Istanbul, Turkey; (D.D.K.); (E.O.)
| | - Ozgul Gok
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey
| |
Collapse
|
4
|
Shi X, Wu K, Liu C, Cao K, Zhang Q, Wu W, Luo C, Huang W. Preliminary investigation into the impact of BPA on osteoblast activity and bone development: In vitro and in vivo models. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 347:123731. [PMID: 38458519 DOI: 10.1016/j.envpol.2024.123731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/24/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Bisphenol A (BPA), an ingredient in consumer products, has been suggested that it can interfere with bone development and maintenance, whereas the molecule mechanism remains unclear. The objective of this study is to investigate the effect of BPA on early bone differentiation and metabolism, and its potential molecule mechanism by employing hFOB1.19 cell as an in vitro model, as well as larval zebrafish as an in vivo model. The in vitro experiments indicated that BPA decreased cell viability, inhibited osteogenic activity (such as ALP, RUNX2), increased ROS production, upregulated transcriptional or protein levels of apoptosis-related molecules (such as Caspase 3, Caspase 9), while suppressed transcriptional or protein levels of pyroptosis-specific markers (TNF-α, TNF-β, IL-1β, ASC, Caspase 1, and GSDMD). Moreover, the evidences from in vivo model demonstrated that exposure to BPA distinctly disrupted pharyngeal cartilage, craniofacial bone development, and retarded bone mineralization. The transcriptional level of bone development-related genes (bmp2, dlx2a, runx2, and sp7), apoptosis-related genes (bcl2), and pyroptosis-related genes (cas1, nlrp3) were significantly altered after treating with BPA in zebrafish larvae. In summary, our study, combining in vitro and in vivo models, confirmed that BPA has detrimental effects on osteoblast activity and bone development. These effects may be due to the promotion of apoptosis, the initiation of oxidative stress, and the inhibition of pyroptosis.
Collapse
Affiliation(s)
- Xiaoling Shi
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Kusheng Wu
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Caixia Liu
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Kexin Cao
- Department of Health Statistics, School of Public Health, Shanxi Medical University, 030001, Taiyuan, China
| | - Qiong Zhang
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Wenying Wu
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Congying Luo
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Wenlong Huang
- Department of Forensic Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
5
|
Nistor M, Rugina D, Diaconeasa Z, Socaciu C, Socaciu MA. Pentacyclic Triterpenoid Phytochemicals with Anticancer Activity: Updated Studies on Mechanisms and Targeted Delivery. Int J Mol Sci 2023; 24:12923. [PMID: 37629103 PMCID: PMC10455110 DOI: 10.3390/ijms241612923] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Pentacyclic triterpenoids (TTs) represent a unique family of phytochemicals with interesting properties and pharmacological effects, with some representatives, such as betulinic acid (BA) and betulin (B), being mainly investigated as potential anticancer molecules. Considering the recent scientific and preclinical investigations, a review of their anticancer mechanisms, structure-related activity, and efficiency improved by their insertion in nanolipid vehicles for targeted delivery is presented. A systematic literature study about their effects on tumor cells in vitro and in vivo, as free molecules or encapsulated in liposomes or nanolipids, is discussed. A special approach is given to liposome-TTs and nanolipid-TTs complexes to be linked to microbubbles, known as contrast agents in ultrasonography. The production of such supramolecular conjugates to deliver the drugs to target cells via sonoporation represents a new scientific and applicative direction to improve TT efficiency, considering that they have limited availability as lipophilic molecules. Relevant and recent examples of in vitro and in vivo studies, as well as the challenges for the next steps towards the application of these complex delivery systems to tumor cells, are discussed, as are the challenges for the next steps towards the application of targeted delivery to tumor cells, opening new directions for innovative nanotechnological solutions.
Collapse
Affiliation(s)
- Madalina Nistor
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Dumitrita Rugina
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Zorita Diaconeasa
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Carmen Socaciu
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Mihai Adrian Socaciu
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
- Department of Radiology, Imaging & Nuclear Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400347 Cluj-Napoca, Romania
| |
Collapse
|
6
|
Adepoju FO, Duru KC, Li E, Kovaleva EG, Tsurkan MV. Pharmacological Potential of Betulin as a Multitarget Compound. Biomolecules 2023; 13:1105. [PMID: 37509141 PMCID: PMC10377123 DOI: 10.3390/biom13071105] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/05/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Betulin is a natural triterpene, usually from birch bark, known for its potential wound-healing properties. Despite having a wide range of pharmacological targets, no studies have proposed betulin as a multitarget compound. Betulin has protective effects against cardiovascular and liver diseases, cancer, diabetes, oxidative stress, and inflammation. It reduces postprandial hyperglycemia by inhibiting α-amylase and α-glucosidase activity, combats tumor cells by inducing apoptosis and inhibiting metastatic proteins, and modulates chronic inflammation by blocking the expression of proinflammatory cytokines via modulation of the NFκB and MAPKs pathways. Given its potential to influence diverse biological networks with high target specificity, it can be hypothesized that betulin may eventually become a new lead for drug development because it can modify a variety of pharmacological targets. The summarized research revealed that the diverse beneficial effects of betulin in various diseases can be attributed, at least in part, to its multitarget anti-inflammatory activity. This review focuses on the natural sources, pharmacokinetics, pharmacological activity of betulin, and the multi-target effects of betulin on signaling pathways such as MAPK, NF-κB, and Nrf2, which are important regulators of the response to oxidative stress and inflammation in the body.
Collapse
Affiliation(s)
- Feyisayo O Adepoju
- Department of Technology for Organic Synthesis, Chemical Technology Institute, Ural Federal University, Mira 19, 620002 Yekaterinburg, Russia
| | - Kingsley C Duru
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ 08854-8021, USA
| | - Erguang Li
- Medical School, Nanjing University, Nanjing, 22 Hankou Road, Nanjing 210093, China
| | - Elena G Kovaleva
- Department of Technology for Organic Synthesis, Chemical Technology Institute, Ural Federal University, Mira 19, 620002 Yekaterinburg, Russia
| | | |
Collapse
|
7
|
Wang S, Wang J, Wang S, Tao R, Yi J, Chen M, Zhao Z. mTOR Signaling Pathway in Bone Diseases Associated with Hyperglycemia. Int J Mol Sci 2023; 24:ijms24119198. [PMID: 37298150 DOI: 10.3390/ijms24119198] [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: 04/03/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 06/12/2023] Open
Abstract
The interplay between bone and glucose metabolism has highlighted hyperglycemia as a potential risk factor for bone diseases. With the increasing prevalence of diabetes mellitus worldwide and its subsequent socioeconomic burden, there is a pressing need to develop a better understanding of the molecular mechanisms involved in hyperglycemia-mediated bone metabolism. The mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that senses extracellular and intracellular signals to regulate numerous biological processes, including cell growth, proliferation, and differentiation. As mounting evidence suggests the involvement of mTOR in diabetic bone disease, we provide a comprehensive review of its effects on bone diseases associated with hyperglycemia. This review summarizes key findings from basic and clinical studies regarding mTOR's roles in regulating bone formation, bone resorption, inflammatory responses, and bone vascularity in hyperglycemia. It also provides valuable insights into future research directions aimed at developing mTOR-targeted therapies for combating diabetic bone diseases.
Collapse
Affiliation(s)
- Shuangcheng Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jiale Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shuangwen Wang
- West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Ran Tao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jianru Yi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Miao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
Ni X, Wu B, Li S, Zhu W, Xu Z, Zhang G, Cui H, Bai Q, Wang J. Equol exerts a protective effect on postmenopausal osteoporosis by upregulating OPG/RANKL pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154509. [PMID: 36288653 DOI: 10.1016/j.phymed.2022.154509] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 10/07/2022] [Accepted: 10/16/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUD Estrogen deficiency is the leading cause of postmenopausal osteoporosis(PMOP) and phytoestrogens soy isoflavones (SI) have been shown to improve PMOP. Equol (Eq), an in vivo metabolite of phytoestrogens soy isoflavones (SI), has a more stable structure and stronger biological activity than its parent compound and has the greatest estrogenic activity. However, there are few studies on the therapeutic effect of Eq on PMOP. PURPOSE To explore the therapeutic effect and mechanisms of Eq on POMP. METHODS Osteoblast-like cells ROS1728 were cultured with different doses of Eq, estradiol (E2), separately. The effect of Eq on the proliferation, apoptosis, cell cycle of osteoblasts were detected by CCK-8 and flow cytometry, and the expression of OPG/RANK/RANKL signaling pathway of osteoblasts was detected by Quantitative real-time PCR (qRT-PCR) and Western blot (WB), and RNA silencing technology were carried out to explore the receptors through which Eq plays a role. Then PMOP rat model was established and treated by Eq or E2 to further verification of the effect and mechanism of Eq on PMOP. RESULT Eq promoted the proliferation and inhibited the apoptosis of osteoblasts and increased the proportion of osteoblasts in the S phase and G2/M phase in a dose-dependent manner. Mechanistically, Eq treatment upregulated the expression of OPG and OPG/RANKL ratio in osteoblasts and this regulatory effect was mainly mediated through the ERβ receptor. Furthermore, in vivo study, Eq improved microstructure and BMD of the femur of PMOP rat model, which imitated the osteoprotective effect of E2. Moreover, the Eq or E2 treatment increased serum levels of Ca, 1,25(OH)2D3, bone Gla-protein(BGP), and Type I procollagen (PC1), and reduced serum levels of phosphorus (P), parathyroid hormone(PTH), pyridinol (PYD), tartrate-resistant acid phosphatase (TRAP) and urinary level of deoxypyridinoline (DPD) in the treatment OVX group compared with the untreated OVX group. Meanwhile, Eq or E2 markedly induced the mRNA and protein expression of OPG and OPG/RANKL ratio. CONCLUSION Eq can combine with ERβ and exert a protective effect on PMOP by upregulating OPG/RANKL pathway.
Collapse
Affiliation(s)
- Xiangmin Ni
- Department of Nutrition, the Second Affiliated Hospital, Army Military Medical University, 400037 Chongqing
| | - Bin Wu
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Army Military Medical University, Chongqing, The sixth medical center of PLA General Hospital, 100142 Beijing
| | - Shuo Li
- Department of Nutrition, the Second Affiliated Hospital, Army Military Medical University, 400037 Chongqing
| | - Wenyi Zhu
- Department of Nutrition, the Second Affiliated Hospital, Army Military Medical University, 400037 Chongqing
| | - Zhe Xu
- Department of Nutrition, the Second Affiliated Hospital, Army Military Medical University, 400037 Chongqing
| | - Guiming Zhang
- Department of Nutrition, the Second Affiliated Hospital, Army Military Medical University, 400037 Chongqing
| | - Hanqiang Cui
- Department of Nutrition, the Second Affiliated Hospital, Army Military Medical University, 400037 Chongqing
| | - Qian Bai
- Department of Nutrition, the Second Affiliated Hospital, Army Military Medical University, 400037 Chongqing
| | - Jian Wang
- Department of Nutrition, the Second Affiliated Hospital, Army Military Medical University, 400037 Chongqing.
| |
Collapse
|
9
|
Vanhie JJ, Kim W, Ek Orloff L, Ngu M, Collao N, De Lisio M. The role of exercise-and high fat diet-induced bone marrow extracellular vesicles in stress hematopoiesis. Front Physiol 2022; 13:1054463. [PMID: 36505084 PMCID: PMC9728614 DOI: 10.3389/fphys.2022.1054463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
Exercise and obesity regulate hematopoiesis, in part through alterations in cellular and soluble components of the bone marrow niche. Extracellular vesicles (EVs) are components of the bone marrow niche that regulate hematopoiesis; however, the role of exercise training or obesity induced EVs in regulating hematopoiesis remains unknown. To address this gap, donor EVs were isolated from control diet-fed, sedentary mice (CON-SED), control diet-fed exercise trained mice (CON-EX), high fat diet-fed, sedentary mice (HFD-SED), and high fat diet-fed, exercise trained mice (HFD-EX) and injected into recipient mice undergoing stress hematopoiesis. Hematopoietic and niche cell populations were quantified, and EV miRNA cargo was evaluated. EV content did not differ between the four groups. Mice receiving HFD-EX EVs had fewer hematopoietic stem cells (HSCs) (p < 0.01), long-term HSC (p < 0.05), multipotent progenitors (p < 0.01), common myeloid progenitors (p<0.01), common lymphoid progenitors (p < 0.01), and granulocyte-macrophage progenitors (p < 0.05), compared to mice receiving HFD-SED EVs. Similarly, mice receiving EX EVs had fewer osteoprogenitor cells compared to SED (p < 0.05) but enhanced mesenchymal stromal cell (MSC) osteogenic differentiation in vitro (p < 0.05) compared to SED EVs. HFD EVs enhanced mesenchymal stromal cell (MSC) adipogenesis in vitro (p < 0.01) compared to CON EVs. HFD-EX EVs had lower microRNA-193 and microRNA-331-5p content, microRNAs implicated in inhibiting osteogenesis and leukemic cell expansion respectively, compared to HFD-SED EVs. The results identify alterations in EV cargo as a novel mechanism by which exercise training alters stress hematopoiesis and the bone marrow niche.
Collapse
Affiliation(s)
- James J. Vanhie
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Wooseok Kim
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Lisa Ek Orloff
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Matthew Ngu
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Nicolas Collao
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Michael De Lisio
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada,*Correspondence: Michael De Lisio,
| |
Collapse
|
10
|
Mizerska-Kowalska M, Sowa S, Donarska B, Płaziński W, Sławińska-Brych A, Tomasik A, Ziarkowska A, Łączkowski KZ, Zdzisińska B. New Borane-Protected Derivatives of α-Aminophosphonous Acid as Anti-Osteosarcoma Agents: ADME Analysis and Molecular Modeling, In Vitro Studies on Anti-Cancer Activities, and NEP Inhibition as a Possible Mechanism of Anti-Proliferative Activity. Int J Mol Sci 2022; 23:ijms23126716. [PMID: 35743158 PMCID: PMC9223658 DOI: 10.3390/ijms23126716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/04/2022] [Accepted: 06/14/2022] [Indexed: 11/18/2022] Open
Abstract
Many organophosphorus compounds (OPs), especially various α-aminophosphonates, exhibit anti-cancer activities. They act, among others, as inhibitors of the proteases implicated in cancerogenesis. Thesetypes of inhibitors weredescribed, e.g., for neutral endopeptidase (NEP) expressed in different cancer cells, including osteosarcoma (OS). The aim of the present study isto evaluate new borane-protected derivatives of phosphonous acid (compounds 1–7) in terms of their drug-likeness properties, anti-osteosarcoma activities in vitro (against HOS and Saos-2 cells), and use as potential NEP inhibitors. The results revealed that all tested compounds exhibited the physicochemical and ADME properties typical for small-molecule drugs. However, compound 4 did not show capability of blood–brain barrier penetration (Lipiński and Veber rules;SwissAdme tool). Moreover, the α-aminophosphonite-boranes (compounds 4–7) exhibited stronger anti-proliferative activity against OS cells than the other phosphonous acid-borane derivatives (compounds 1–3),especially regarding HOS cells (MTT assay). The most promising compounds 4 and 6 induced apoptosis through the activation of caspase 3 and/or cell cycle arrest at the G2 phase (flow cytometry). Compound 4 inhibited the migration and invasiveness of highly aggressive HOS cells (wound/transwell and BME-coated transwell assays, respectively). Additionally, compound 4 and, to a lesser extent, compound 6 inhibited NEP activity (fluorometric assay). This activity of compound 4 was involved in its anti-proliferative potential (BrdU assay). The present study shows that compound 4 can be considered a potential anti-osteosarcoma agent and a scaffold for the development of new NEP inhibitors.
Collapse
Affiliation(s)
- Magdalena Mizerska-Kowalska
- Department of Virology and Immunology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19 Street, 20-033 Lublin, Poland; (A.T.); (A.Z.); (B.Z.)
- Correspondence:
| | - Sylwia Sowa
- Faculty of Chemistry, Department of Organic Chemistry, Maria Curie-Skłodowska University, Gliniana 33 Street, 20-614 Lublin, Poland;
| | - Beata Donarska
- Faculty of Pharmacy, Collegium Medicum, Department of Chemical Technology and Pharmaceuticals, Nicolaus Copernicus University, Jurasza 2 Street, 85-089 Bydgoszcz, Poland; (B.D.); (K.Z.Ł.)
| | - Wojciech Płaziński
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8 Street, 30-239 Cracow, Poland;
- Department of Biopharmacy, Faculty of Pharmacy, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland
| | - Adrianna Sławińska-Brych
- Department of Cell Biology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19 Street, 20-033 Lublin, Poland;
| | - Aleksandra Tomasik
- Department of Virology and Immunology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19 Street, 20-033 Lublin, Poland; (A.T.); (A.Z.); (B.Z.)
| | - Anna Ziarkowska
- Department of Virology and Immunology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19 Street, 20-033 Lublin, Poland; (A.T.); (A.Z.); (B.Z.)
| | - Krzysztof Z. Łączkowski
- Faculty of Pharmacy, Collegium Medicum, Department of Chemical Technology and Pharmaceuticals, Nicolaus Copernicus University, Jurasza 2 Street, 85-089 Bydgoszcz, Poland; (B.D.); (K.Z.Ł.)
| | - Barbara Zdzisińska
- Department of Virology and Immunology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19 Street, 20-033 Lublin, Poland; (A.T.); (A.Z.); (B.Z.)
| |
Collapse
|
11
|
Yang K, Cao F, Xue Y, Tao L, Zhu Y. Three Classes of Antioxidant Defense Systems and the Development of Postmenopausal Osteoporosis. Front Physiol 2022; 13:840293. [PMID: 35309045 PMCID: PMC8927967 DOI: 10.3389/fphys.2022.840293] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/25/2022] [Indexed: 01/04/2023] Open
Abstract
Osteoporosis is a common bone imbalance disease that threatens the health of postmenopausal women. Estrogen deficiency accelerates the aging of women. Oxidative stress damage is regarded as the main pathogenesis of postmenopausal osteoporosis. The accumulation of reactive oxygen species in the bone microenvironment plays a role in osteoblast and osteoclast apoptosis. Improving the oxidative state is essential for the prevention and treatment of postmenopausal osteoporosis. There are three classes of antioxidant defense systems in the body to eliminate free radicals and peroxides including antioxidant substances, antioxidant enzymes, and repair enzymes. In our review, we demonstrated the mechanism of antioxidants and their effect on bone metabolism in detail. We concluded that glutathione/oxidized glutathione (GSH/GSSG) conversion involved the PI3K/Akt-Nrf2/HO-1 signaling pathway and that the antioxidant enzyme-mediated mitochondrial apoptosis pathway of osteoblasts was necessary for the development of postmenopausal osteoporosis. Since the current therapeutic effects of targeting bone cells are not significant, improving the systemic peroxidation state and then regulating bone homeostasis will be a new method for the treatment of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Keda Yang
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
| | - Fangming Cao
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
| | - Yuchuan Xue
- The First Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Lin Tao
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
- *Correspondence: Lin Tao,
| | - Yue Zhu
- Department of Orthopedics, First Hospital of China Medical University, Shenyang, China
- Yue Zhu,
| |
Collapse
|
12
|
Drasar PB, Khripach VA. Terpene Research Is Providing New Inspiration for Scientists. Molecules 2021; 26:molecules26185480. [PMID: 34576951 PMCID: PMC8465594 DOI: 10.3390/molecules26185480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Affiliation(s)
- Pavel B. Drasar
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Technicka 5, 166 28 Prague, Czech Republic
- Correspondence:
| | - Vladimir A. Khripach
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, 5/2 Academician V. F. Kuprevich Street, BY-220141 Minsk, Belarus;
| |
Collapse
|
13
|
adipoSIGHT in Therapeutic Response: Consequences in Osteosarcoma Treatment. Bioengineering (Basel) 2021; 8:bioengineering8060083. [PMID: 34200614 PMCID: PMC8229256 DOI: 10.3390/bioengineering8060083] [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] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/02/2022] Open
Abstract
Chemotherapeutic resistance is a major problem in effective cancer treatment. Cancer cells engage various cells or mechanisms to resist anti-cancer therapeutics, which results in metastasis and the recurrence of disease. Considering the cellular heterogeneity of cancer stroma, the involvement of stem cells is reported to affect the proliferation and metastasis of osteosarcoma. Hence, the duo (osteosarcoma: Saos 2 and human adipose-derived stem cells: ASCs) is co-cultured in present study to investigate the therapeutic response using a nonadherent, concave surface. Staining with a cell tracker allows real-time microscopic monitoring of the cell arrangement within the sphere. Cell–cell interaction is investigated by means of E-cadherin expression. Comparatively high expression of E-cadherin and compact organization is observed in heterotypic tumorspheres (Saos 2–ASCs) compared to homotypic ones (ASCs), limiting the infiltration of chemotherapeutic compound doxorubicin into the heterotypic tumorsphere, which in turn protects cells from the toxic effect of the chemotherapeutic. In addition, genes known to be associated with drug resistance, such as SOX2, OCT4, and CD44 are overexpressed in heterotypic tumorspheres post-chemotherapy, indicating that the duo collectively repels the effect of doxorubicin. The interaction between ASCs and Saos 2 in the present study points toward the growing oncological risk of using ASC-based regenerative therapy in cancer patients and warrants further investigation.
Collapse
|
14
|
Wang JK, Çimenoğlu Ç, Cheam NMJ, Hu X, Tay CY. Sustainable aquaculture side-streams derived hybrid biocomposite for bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112104. [PMID: 34082928 DOI: 10.1016/j.msec.2021.112104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/23/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023]
Abstract
Despite being a rich source of bioactive compounds, the current exploitation of aquatic biomass is insufficient. Majority of the aquaculture industry side-streams are currently used for low-value purposes such as animal feed or composting material, with low economical returns. To maximize resource reuse and minimize waste generation, valorization efforts should be augmented with the aim to produce high-value products. Herein, we present a novel aquaculture wastes-derived multi-scale osteoconductive hybrid biocomposite that is composed of chemically crosslinked American bullfrog (Rana catesbeiana) skin-derived type I tropocollagen nanofibrils (~22.3 nm) network and functionalized with micronized (~1.6 μm) single-phase hydroxyapatite (HA) from discarded snakehead (Channa micropeltes) fish scales. The bioengineered construct is biocompatible, highly porous (>90%), and exhibits excellent osteoconductive properties, as indicated by robust adhesion and proliferation of human fetal osteoblastic 1.19 cell line (hFOB 1.19). Furthermore, increased expression level of osteo-related ALPL and BGLAP mRNA transcripts, as well as enhanced osteocalcin immunoreactivity and increasing Alizarin red S staining coverage on the hybrid biocomposite was observed over 21 days of culture. Collectively, the devised "waste-to-resource" platform represents a sustainable waste valorization strategy that is amendable for advanced bone repair and regeneration applications.
Collapse
Affiliation(s)
- Jun Kit Wang
- School of Materials Science and Engineering, Nanyang Technological University Singapore, N4.1, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Çiğdem Çimenoğlu
- School of Materials Science and Engineering, Nanyang Technological University Singapore, N4.1, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Nicole Mein Ji Cheam
- School of Materials Science and Engineering, Nanyang Technological University Singapore, N4.1, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Xiao Hu
- School of Materials Science and Engineering, Nanyang Technological University Singapore, N4.1, 50 Nanyang Avenue, Singapore 639798, Singapore; Environmental Chemistry and Materials Centre, Nanyang Environment & Water Research Institute, 1 CleanTech Loop, CleanTech One, Singapore 637141, Singapore
| | - Chor Yong Tay
- School of Materials Science and Engineering, Nanyang Technological University Singapore, N4.1, 50 Nanyang Avenue, Singapore 639798, Singapore; Environmental Chemistry and Materials Centre, Nanyang Environment & Water Research Institute, 1 CleanTech Loop, CleanTech One, Singapore 637141, Singapore; School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore; Energy Research Institute, Nanyang Technological University Singapore, 50 Nanyang Drive, Singapore 637553, Singapore.
| |
Collapse
|
15
|
Ortega MA, Asúnsolo Á, Pekarek L, Alvarez-Mon MA, Delforge A, Sáez MA, Coca S, Sainz F, Mon MÁ, Buján J, García-Honduvilla N. Histopathological study of JNK in venous wall of patients with chronic venous insufficiency related to osteogenesis process. Int J Med Sci 2021; 18:1921-1934. [PMID: 33850461 PMCID: PMC8040408 DOI: 10.7150/ijms.54052] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic venous insufficiency (CVI) is one of the most common vascular pathologies worldwide. One of the risk factors for the development of CVI is aging, which is why it is related to senile changes. The main trigger of the changes that occur in the venous walls in CVI is blood flow reflux, which produces increased hydrostatic pressure, leading to valve incompetence. The cellular response is one of the fundamental processes in vascular diseases, causing the activation of cell signalling pathways such as c-Jun N-terminal kinase (JNK). Metabolic changes and calcifications occur in vascular pathology as a result of pathophysiological processes. The aim of this study was to determine the expression of JNK in venous disease and its relationship with the role played by the molecules involved in the osteogenic processes in venous tissue calcification. This was a cross-sectional study that analyzed the greater saphenous vein wall in 110 patients with (R) and without venous reflux (NR), classified according to age. Histopathological techniques were used and protein expression was analysed using immunohistochemistry techniques for JNK and markers of osteogenesis (RUNX2, osteocalcin (OCN), osteopontin (OPN)). Significantly increased JNK, RUNX2, OCN, OPN and pigment epithelium-derived factor (PEDF) protein expression and the presence of osseous metaplasia and amorphous calcification were observed in younger patients (<50 years) with venous reflux. This study shows for the first time the existence of an osteogenesis process related to the expression of JNK in the venous wall.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Ángel Asúnsolo
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Leonel Pekarek
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Miguel A Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Arnaud Delforge
- UFR of pharmacy, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Miguel A Sáez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, Spain
| | - Santiago Coca
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Felipe Sainz
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Angiology and Vascular Surgery Service, Central University Hospital of Defence-UAH Madrid, Spain
| | - Melchor Álvarez- Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service and Internal Medicine, University Hospital Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Julia Buján
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
| |
Collapse
|
16
|
MicroRNA-409-3p promotes osteoblastic differentiation via activation of Wnt/β-catenin signaling pathway by targeting SCAI. Biosci Rep 2021; 41:227142. [PMID: 33274736 PMCID: PMC7791548 DOI: 10.1042/bsr20201902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/18/2020] [Accepted: 12/03/2020] [Indexed: 11/17/2022] Open
Abstract
Osteogenic differentiation is an important process of new bone formation, microRNA-409-3p (miR-409-3p) has been reported to be up-regulated in the osteogenic differentiation of human bone marrow mesenchymal stem cells (MSCs). The present study aimed to investigate the regulatory effect of miR-409-3p on osteogenic differentiation of MSCs and its molecular mechanism. The expression of miR-409-3p in osteoblast (human skull osteoblast, HCO) and bone marrow-derived MSCs (MSC-A, MSC-B, MSC-U) were detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The binding of miR-409-3p to suppressor of cancer cell invasion (SCAI) in MSC-B was investigated by performing a dual-luciferase reporter gene assay. MSC-B was selected to transfect with miR-409-3p analog/complementary sequence (cs), miR-409-3p analog + SCAI and miR-409-3p cs + small interfering (si)-SCAI, as well as control, respectively. The alkaline phosphatase (ALP) activity, Alizarin Red staining, and the expression of osteogenic markers (ALP, osteocalcin (OCN), osteopontin (OPN), runt-related transcription factor 2 (RUNX2)) in MSC-B during osteoblastic differentiation were tested by RT-qPCR and Western blotting, respectively. Additionally, the Wnt/β-catenin pathway was inhibited by dickkopf-related protein 1 (DKK-1) to get the roles of miR-409-3p during the osteoblastic differentiation of MSC-B when transfected with miR-409-3p analog. The expression of miR-409-3p in HCO was higher than that in these three MSCs and showed an increasing time-dependent trend on the 0 and 21st day of osteoblastic differentiation. MiR-409-3p directly regulated SCAI by targeting SCAI 3′UTR. Further, miR-409-3p suppressed SCAI expression, but SCAI up-regulation suppressed the osteoblastic differentiation, as well as reduced the relative mRNA/protein expression of Wnt/β-catenin signaling pathway-related genes (Axis inhibition protein 1 (AXIN1), β-catenin, Lymphoid Enhancer Binding Factor 1, Cellular-myelocytomatosis (c-myc) and cyclin D1). Importantly, disruption of Wnt signaling also blocked miR-409-3p induced osteoblastic differentiation of MSCs. Therefore, miR-409-3p promotes osteoblastic differentiation through the activation of the Wnt/β-catenin pathway by down-regulating SCAI expression.
Collapse
|
17
|
Careta O, Salicio-Paz A, Pellicer E, Ibáñez E, Fornell J, García-Lecina E, Sort J, Nogués C. Electroless Palladium-Coated Polymer Scaffolds for Electrical Stimulation of Osteoblast-Like Saos-2 Cells. Int J Mol Sci 2021; 22:E528. [PMID: 33430266 PMCID: PMC7825691 DOI: 10.3390/ijms22020528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 12/29/2020] [Accepted: 01/03/2021] [Indexed: 11/16/2022] Open
Abstract
Three-dimensional porous scaffolds offer some advantages over conventional treatments for bone tissue engineering. Amongst all non-bioresorbable scaffolds, biocompatible metallic scaffolds are preferred over ceramic and polymeric scaffolds, as they can be used as electrodes with different electric field intensities (or voltages) for electric stimulation (ES). In the present work we have used a palladium-coated polymeric scaffold, generated by electroless deposition, as a bipolar electrode to electrically stimulate human osteoblast-like Saos-2 cells. Cells grown on palladium-coated polyurethane foams under ES presented higher proliferation than cells grown on foams without ES for up to 14 days. In addition, cells grown in both conditions were well adhered, with a flat appearance and a typical actin cytoskeleton distribution. However, after 28 days in culture, cells without ES were filling the entire structure, while cells under ES appeared rounded and not well adhered, a sign of cell death onset. Regarding osteoblast differentiation, ES seems to enhance the expression of early expressed genes. The results suggest that palladium-coated polyurethane foams may be good candidates for osteoblast scaffolds and demonstrate that ES enhances osteoblast proliferation up to 14 days and upregulate expression genes related to extracellular matrix formation.
Collapse
Affiliation(s)
- Oriol Careta
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain; (O.C.); (E.I.)
| | - Asier Salicio-Paz
- CIDETEC, Basque Research and Technology Alliance (BRTA), Paseo Miramón 196, E-20014 Donostia-San Sebastián, Spain; (A.S.-P.); (E.G.-L.)
| | - Eva Pellicer
- Departament de Física, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain; (E.P.); (J.S.)
| | - Elena Ibáñez
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain; (O.C.); (E.I.)
| | - Jordina Fornell
- Departament de Física, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain; (E.P.); (J.S.)
| | - Eva García-Lecina
- CIDETEC, Basque Research and Technology Alliance (BRTA), Paseo Miramón 196, E-20014 Donostia-San Sebastián, Spain; (A.S.-P.); (E.G.-L.)
| | - Jordi Sort
- Departament de Física, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain; (E.P.); (J.S.)
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys 23, E-08180 Barcelona, Spain
| | - Carme Nogués
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain; (O.C.); (E.I.)
| |
Collapse
|
18
|
MASSUNARI L, RABELO RL, LEITE ML, SOARES DG, ANOVAZZI G, COSTA CAS, DUQUE C. Dose- and time-dependent effects of taxifolin on viability and mineralization markers of osteoblast-like cells. Braz Oral Res 2021; 35:e140. [DOI: 10.1590/1807-3107bor-2021.vol35.0140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 07/13/2021] [Indexed: 11/22/2022] Open
|
19
|
Zhu WQ, Ming PP, Zhang SM, Qiu J. Role of MAPK/JNK signaling pathway on the regulation of biological behaviors of MC3T3‑E1 osteoblasts under titanium ion exposure. Mol Med Rep 2020; 22:4792-4800. [PMID: 33173964 PMCID: PMC7646925 DOI: 10.3892/mmr.2020.11575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
The oral cavity is a complex environment that is constantly undergoing remodeling. This provides a favorable electrolytic aqueous condition, which causes the corrosion of titanium implants and the release of titanium (Ti) ions. The accumulation of Ti ions in the peri-implant tissues may affect the osteogenesis process. Therefore, the present study aimed to investigate the possible effects of Ti ions on osteoblast physiology and its underlying mechanism, specifically the MAPK/JNK signaling pathway. In the present study, MC3T3-E1 osteoblasts were cultured the medium containing 10 ppm Ti ions. Confocal laser scanning microscopy was used to analyze cell morphology and adhesion. Alkaline phosphatase (ALP) activity assay and western blotting were performed to evaluate the expression of proteins associated with osteogenesis such as Runx2 and Osterix. Nuclear translocation of JNK, a key factor of the MAPK signaling pathway, was visualized and analyzed using immunofluorescence staining. The results showed that 10 ppm Ti ions exerted negative effects on the biological behaviors of MC3T3-E1 cells, which exhibited reduced adhesion, ALP activity and osteogenic differentiation. It was also found that 10 ppm Ti ions activated the MAPK/JNK signaling pathway by promoting the nuclear translocation of JNK via phosphorylation. In addition, the inhibitory effects of 10 ppm Ti ions on MC3T3-E1 cells was found to be reversed by the JNK inhibitor SP600125. In conclusion, the preset study suggests that the MAPK/JNK signaling pathway serves a key role in the molecular mechanism underlying the changes in osteoblast behavior following Ti ion exposure. These findings may serve as a valuable reference point for the further in-depth exploration of peri-implant bone loss.
Collapse
Affiliation(s)
- Wen-Qing Zhu
- Department of Oral Implantology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Pan-Pan Ming
- Department of Stomatology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Song-Mei Zhang
- Department of General Dentistry, Eastman Institute for Oral Health, University of Rochester, Rochester, NY 14620, USA
| | - Jing Qiu
- Department of Oral Implantology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
20
|
Lin YC, Chen HY, Hsieh CP, Huang YF, Chang IL. Betulin inhibits mTOR and induces autophagy to promote apoptosis in human osteosarcoma cell lines. ENVIRONMENTAL TOXICOLOGY 2020; 35:879-887. [PMID: 32190974 DOI: 10.1002/tox.22924] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/18/2020] [Accepted: 03/06/2020] [Indexed: 05/20/2023]
Abstract
Betulin is a lupane type pentacyclic triterpenoid, and commonly found in the bark of birch trees. It displays various pharmacological properties, such as antibacterial, anti-inflammation, antitumor, and antiviral. In this report, we attempted to investigate the anti-proliferative and pro-apoptotic effects of betulin on osteosarcoma cell lines. Our results revealed that betulin significantly decreased cell viability and colony formation in osteosarcoma cell lines. Dose-dependent induction of Annexin V positive cells, activated caspase 8, activated caspase 9, activated caspase 3, and the cleavage of poly (ADP-ribose) polymerase were observed after the treatment with betulin, indicating betulin induces apoptosis in osteosarcoma cell lines. mTOR has been identified as a key modulator of autophagy in response to different stresses. In this study, we found that the treatment with betulin suppressed the activation of mTOR, and increased the level of LC 3-II, the autophagy marker, in osteosarcoma cell lines. Co-administration of the autophagy inhibitor chloroquine significantly rescued the cell viability and the clonogenic activity in betulin-treated osteosarcoma cell lines. Our data showed that betulin induced autophagy, and the up-regulated autophagy positively contributed to the apoptosis. Taken together, our findings suggested that betulin may serve as a promising anti-proliferative agent for treating osteosarcoma.
Collapse
Affiliation(s)
- Yung-Chi Lin
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsuan-Ying Chen
- Orthopedics & Sports Medicine Laboratory, Changhua Christian Hospital, Changhua, Taiwan
| | - Cheng-Pu Hsieh
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan
- Orthopedics & Sports Medicine Laboratory, Changhua Christian Hospital, Changhua, Taiwan
| | - Yi-Fu Huang
- Orthopedics & Sports Medicine Laboratory, Changhua Christian Hospital, Changhua, Taiwan
| | - Ing-Lin Chang
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
21
|
Lin YT, Mao YW, Imtiyaz Z, Chiou WF, Lee MH. Comprehensive LC-MS/MS-based phytochemical perspectives and osteogenic effects of Uraria crinita. Food Funct 2020; 11:5420-5431. [PMID: 32475999 DOI: 10.1039/d0fo00782j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Osteogenesis plays a vital role in the maintenance of bone health. Imbalances in osteogenesis influence the onset of several bone loss-associated diseases. The intake of Uraria crinita (Fabaceae) through dietary supplements is advised for childhood bone dysplasia. This botanical provides edible tonics and detoxifiers, and is also used as a folk beverage. We evaluated the osteogenic effects of a 50% ethanol extract of the root of U. crinita on primary human osteoblasts (HObs) and initiated a novel comprehensive phytochemical strategy using liquid chromatography-tandem mass spectrometry (LC-MS/MS) for quality control of this functional food. Two isoflavones, genistein (5) and 5,7-dihydroxy-3',5'-dihydroxyisoflavone (6), increased the alkaline phosphatase activity (differentiation stage); the flavone glycoside vitexin (1), and the phenolic acid salicylic acid (2) enhanced the mineralization (mature stage). The isoflavone 2'-hydroxygenistein (4) possessed high osteogenic potential among the isolated compounds in HObs. It promoted osteogenesis-related stages and upregulated the gene expressions in a dose-dependent manner. The major compounds in the active fraction were quantitatively analyzed via phytochemical fingerprint detection. These LC-MS/MS-based phytochemical perspectives can act as reference standards in developing food supplements from U. crinita.
Collapse
Affiliation(s)
- Yi-Tzu Lin
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yi-Wen Mao
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| | - Zuha Imtiyaz
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| | - Wen-Fei Chiou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan.
| | - Mei-Hsien Lee
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan. and Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan and Center for Reproductive Medicine & Sciences, Taipei Medical University Hospital, Taipei 11031, Taiwan
| |
Collapse
|
22
|
Wang S, Qiu J, Guo A, Ren R, He W, Liu S, Liu Y. Nanoscale perfluorocarbon expediates bone fracture healing through selectively activating osteoblastic differentiation and functions. J Nanobiotechnology 2020; 18:84. [PMID: 32493334 PMCID: PMC7271395 DOI: 10.1186/s12951-020-00641-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/25/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND RATIONALE Fracture incidence increases with ageing and other contingencies. However, the strategy of accelerating fracture repair in clinical therapeutics remain a huge challenge due to its complexity and a long-lasting period. The emergence of nano-based drug delivery systems provides a highly efficient, targeted and controllable drug release at the diseased site. Thus far, fairly limited studies have been carried out using nanomedicines for the bone repair applications. Perfluorocarbon (PFC), FDA-approved clinical drug, is received increasing attention in nanomedicine due to its favorable chemical and biologic inertness, great biocompatibility, high oxygen affinity and serum-resistant capability. In the premise, the purpose of the current study is to prepare nano-sized PFC materials and to evaluate their advisable effects on promoting bone fracture repair. RESULTS Our data unveiled that nano-PFC significantly enhanced the fracture repair in the rabbit model with radial fractures, as evidenced by increased soft callus formation, collagen synthesis and accumulation of beneficial cytokines (e.g., vascular endothelial growth factor (VEGF), matrix metalloprotein 9 (MMP-9) and osteocalcin). Mechanistic studies unraveled that nano-PFC functioned to target osteoblasts by stimulating their differentiation and activities in bone formation, leading to accelerated bone remodeling in the fractured zones. Otherwise, osteoclasts were not affected upon nano-PFC treatment, ruling out the potential target of nano-PFC on osteoclasts and their progenitors. CONCLUSIONS These results suggest that nano-PFC provides a potential perspective for selectively targeting osteoblast cell and facilitating callus generation. This study opens up a new avenue for nano-PFC as a promising agent in therapeutics to shorten healing time in treating bone fracture.
Collapse
Affiliation(s)
- Shunhao Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 8 Shuangqing Road, Haidian District, Beijing, 100085, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiahuang Qiu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 8 Shuangqing Road, Haidian District, Beijing, 100085, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Anyi Guo
- Beijing Jishuitan Hospital, The 4th Clinical Hospital of Peking University Health Science Center, No. 31 East Street, Xinjiekou, Xicheng District, Beijing, 100035, China
| | - Ruanzhong Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 8 Shuangqing Road, Haidian District, Beijing, 100085, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei He
- Beijing Jishuitan Hospital, The 4th Clinical Hospital of Peking University Health Science Center, No. 31 East Street, Xinjiekou, Xicheng District, Beijing, 100035, China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 8 Shuangqing Road, Haidian District, Beijing, 100085, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yajun Liu
- Beijing Jishuitan Hospital, The 4th Clinical Hospital of Peking University Health Science Center, No. 31 East Street, Xinjiekou, Xicheng District, Beijing, 100035, China.
| |
Collapse
|