1
|
Rahaman SH, Bodhak S, Balla VK, Bhattacharya D. Role of in-situ electrical stimulation on early-stage mineralization and in-vitro osteogenesis of electroactive bioactive glass composites. BIOMATERIALS ADVANCES 2025; 166:214062. [PMID: 39406157 DOI: 10.1016/j.bioadv.2024.214062] [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: 08/13/2024] [Accepted: 10/07/2024] [Indexed: 11/13/2024]
Abstract
Bioactive glass (BAG) has emerged as an effective bone graft substitute due to its diverse qualities of biocompatibility, bioactivity, osteoblast adhesion and enhanced revascularization. However, inferior osteogenic capacity of BAG compared to autologous bone grafts continues to limiting it's wide-spread clinical applications towards repairing of bone fractures and healing. In this study, we have fabricated BAG composites with 0.5 to 2 wt% bismuth ferrite (BF, a multiferroic material) with an aim to generate in-situ electrical charges pertinent to early-stage bone regeneration thus mimicking natural bone, which is a piezoelectric material. The fabricated BAG composites were characterised in terms of microstructures, phase analysis, remanent polarization, wettability and subsequently evaluated for in vitro cell proliferation and osteogenesis with and without magnetic field exposure (200 mT, 30 min./day). Pre-osteoblast cells from mice (MC3T3-E1) seeded on these composites exhibited excellent cell growth without any cytotoxicity, which is further supported by FITC/DAPI staining and a live/dead assay. The results of Alizarin Red S assay and increased levels of Alkaline Phosphatase (ALP) activity, at 21 days of culture, suggest that the BAG-BF composites promote in vitro osteogenic differentiation of pre-osteoblast cells. The enhanced osteogenesis of BAG-BF composites was also confirmed through qRT-PCR analysis, which showed rapid upregulation of osteoblastogenic specific genes namely RunX-2, Collagen-1, Bone Sialo Protein, and ALP after 21 days. Additionally, the osteogenic differentiation was assessed by the Western Blot technique, which revealed significantly higher band intensity of osteogenic markers in BAG-1.5 BF and BAG-2 BF composites than pure BAG. These findings clearly demonstrate that in-situ electrical stimulation and osteoconductive capacity of BF reinforced BAG composites have positive impact on osteoblast cell development, bone formation, and healing.
Collapse
Affiliation(s)
- Sk Hasanur Rahaman
- Bioceramics and Coating Division, CSIR-Central Glass and Ceramic Research Institute, 196, Raja S.C Mullick Road, Jadavpur, Kolkata 700 032, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Subhadip Bodhak
- Bioceramics and Coating Division, CSIR-Central Glass and Ceramic Research Institute, 196, Raja S.C Mullick Road, Jadavpur, Kolkata 700 032, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Vamsi Krishna Balla
- Bioceramics and Coating Division, CSIR-Central Glass and Ceramic Research Institute, 196, Raja S.C Mullick Road, Jadavpur, Kolkata 700 032, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Dipten Bhattacharya
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Advanced Mechanical and Materials Characterization Division, CSIR-Central Glass and Ceramic Research Institute, 196, Raja S.C Mullick Road, Jadavpur, Kolkata 700 032, India
| |
Collapse
|
2
|
Sohrabi M, Hesaraki S, Shahrezaee M, Shams-Khorasani A. The release behavior and in vitro osteogenesis of quercetin-loaded bioactive glass/hyaluronic acid/sodium alginate nanocomposite paste. Int J Biol Macromol 2024; 280:136094. [PMID: 39343279 DOI: 10.1016/j.ijbiomac.2024.136094] [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: 02/03/2024] [Revised: 09/11/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Injectable pastes based on bioactive compounds and natural polymers are of interest in non-invasive bone surgeries. Several quantities of quercetin (100, 150, and 200 μM) were added to a sol-gel derived mesoporous bioactive glass. Injectable pastes based on quercetin-loaded bioactive glass, sodium alginate, and hyaluronic acid were prepared. Aggregated nanoparticles of bioactive glass and quercetin-loaded bioactive glass with mesoporous morphologies were confirmed by TEM and BET techniques. The quercetin release study was assessed in phosphate-buffered solution medium over 200 h and the obtained data were fitted by different eqs. A sustained release of quercetin was found, in which a better regression coefficient was achieved using Weibull equation. Human-derived mesenchymal stem cells were utilized to determine alkaline phosphatase activity and bone-related protein expression by western blotting and real-time PCR evaluations. Quercetin-loaded pastes increased the levels of alkaline phosphatase activity and the expression of Collagen-1, Osteopontin, Osteocalcin, and Runx2 proteins in a concentration-dependent manner. Due to the mesoporous architecture and high specific surface area of bioactive glass, the paste made of these particles and sodium alginate/hyaluronic acid macromolecules is appropriate matrix for quercetin release, resulting in promoted osteogenesis. The further in vivo studies can support the osteogenesis capacity of the quercetin-loaded paste.
Collapse
Affiliation(s)
- Mehri Sohrabi
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Alborz, Iran.
| | - Saeed Hesaraki
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Alborz, Iran.
| | | | - Alireza Shams-Khorasani
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Alborz, Iran
| |
Collapse
|
3
|
Liu SF, Kucherenko MM, Sang P, Li Q, Yao J, Nambiar Veetil N, Gransar T, Alesutan I, Voelkl J, Salinas G, Grune J, Simmons S, Knosalla C, Kuebler WM. RUNX2 is stabilised by TAZ and drives pulmonary artery calcification and lung vascular remodelling in pulmonary hypertension due to left heart disease. Eur Respir J 2024; 64:2300844. [PMID: 39542509 DOI: 10.1183/13993003.00844-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/13/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Calcification is common in chronic vascular disease, yet its role in pulmonary hypertension due to left heart disease is unknown. Here, we probed for the role of runt-related transcription factor-2 (RUNX2), a master transcription factor in osteogenesis, and its regulation by the HIPPO pathway transcriptional coactivator with PDZ-binding motif (TAZ) in the osteogenic reprogramming of pulmonary artery smooth muscle cells and vascular calcification in patients with pulmonary hypertension due to left heart disease. We similarly examined its role using an established rat model of pulmonary hypertension due to left heart disease induced by supracoronary aortic banding. METHODS Pulmonary artery samples were collected from patients and rats with pulmonary hypertension due to left heart disease. Genome-wide RNA sequencing was performed, and pulmonary artery calcification assessed. Osteogenic signalling via TAZ and RUNX2 was delineated by protein biochemistry. In vivo, the therapeutic potential of RUNX2 or TAZ inhibition by CADD522 or verteporfin was tested in the rat model. RESULTS Gene ontology term analysis identified significant enrichment in ossification and osteoblast differentiation genes, including RUNX2, in pulmonary arteries of patients and lungs of rats with pulmonary hypertension due to left heart disease. Pulmonary artery calcification was evident in both patients and rats. Both TAZ and RUNX2 were upregulated and activated in pulmonary artery smooth muscle cells of patients and rats. Co-immunoprecipitation revealed a direct interaction of RUNX2 with TAZ in pulmonary artery smooth muscle cells. TAZ inhibition or knockdown decreased RUNX2 abundance due to accelerated RUNX2 protein degradation rather than reduced de novo synthesis. Inhibition of either TAZ or RUNX2 attenuated pulmonary artery calcification, distal lung vascular remodelling and pulmonary hypertension development in the rat model. CONCLUSION Pulmonary hypertension due to left heart disease is associated with pulmonary artery calcification that is driven by TAZ-dependent stabilisation of RUNX2, causing osteogenic reprogramming of pulmonary artery smooth muscle cells. The TAZ-RUNX2 axis may present a therapeutic target in pulmonary hypertension due to left heart disease.
Collapse
Affiliation(s)
- Shao-Fei Liu
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- These authors contributed equally to the study
| | - Mariya M Kucherenko
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- These authors contributed equally to the study
| | - Pengchao Sang
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Qiuhua Li
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Juquan Yao
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Netra Nambiar Veetil
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
| | - Tara Gransar
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Ioana Alesutan
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Jakob Voelkl
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriela Salinas
- NGS - Integrative Genomics Core Unit (NIG), Institute of Pathology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Jana Grune
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Szandor Simmons
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
| | - Christoph Knosalla
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- These authors share the last authorship
| | - Wolfgang M Kuebler
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- DZL (German Centre for Lung Research), partner site Berlin, Berlin, Germany
- The Keenan Research Centre for Biomedical Science at St. Michael's, Toronto, ON, Canada
- Departements of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
- These authors share the last authorship
| |
Collapse
|
4
|
Arab S, Bahraminasab M, Asgharzade S, Doostmohammadi A, Zadeh ZK, Nooshabadi VT. On the osteogenic differentiation of dental pulp stem cells by a fabricated porous nano-hydroxyapatite substrate loaded with sodium fluoride. BMC Oral Health 2024; 24:1218. [PMID: 39402484 PMCID: PMC11476061 DOI: 10.1186/s12903-024-04987-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
In the present study, nano-hydroxyapatite (n-HA) powder was extracted from carp bone waste to fabricate porous n-HA substrates by a molding and sintering process. Subsequently, the substrates were loaded with different amounts of sodium fluoride (NaF) through immersion in NaF suspensions for 10, 7.5, and 5 min. The NaF-loaded n-HA substrates were then examined for their structural and physical properties, chemical bonds, loading and release profile, pH changes, cytotoxicity, and osteogenic effect on dental pulp stem cells (DPSCs) at the level of RNA and protein expression. The results showed that the n-HA substrates were porous (> 40% porosity) and had rough surfaces. The NaF could be successfully loaded on the substrates, which was 6.43, 4.50, and 1.47 mg, respectively for n-HA substrates with immersion times of 10, 7.5, and 5 min in the NaF suspensions. It was observed that the NaF release rate was rather fast during the first 24 h in all groups (39.06%, 36.43%, and 39.57% for 10, 7.5, and 5 min, respectively), and decreased dramatically after that, indicating a slow detachment of NaF. Furthermore, the pH of the medium related to all materials was changed during the first 4 days of immersion (from 7.38 to pH of about 7.85, 7.84, 7.63, and 7.66 for C0, C5, C7.5, and C10, respectively). The pH of media associated with the C7.5, and C10 increased up to 4 days and remained relatively constant until day 14 (pH = 7.6). The results of the cytotoxicity assay rejected any toxicity of the fabricated NaF-loaded n-HA substrates on DPSCs, and the cells could adhere to their surfaces with enlarged morphology. The results showed no effect on the osteogenic differentiation at the protein level. Nevertheless, this effect was observed at the gene level.
Collapse
Affiliation(s)
- Samaneh Arab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Marjan Bahraminasab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | - Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ali Doostmohammadi
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Zahra Khatib Zadeh
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
5
|
Roth DM, Piña JO, MacPherson M, Budden C, Graf D. Physiology and Clinical Manifestations of Pathologic Cranial Suture Widening. Cleft Palate Craniofac J 2024; 61:1750-1759. [PMID: 37271984 PMCID: PMC11468227 DOI: 10.1177/10556656231178438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023] Open
Abstract
Cranial sutures are complex structures integrating mechanical forces with osteogenesis which are often affected in craniofacial syndromes. While premature fusion is frequently described, rare pathological widening of cranial sutures is a comparatively understudied phenomenon. This narrative review aims to bring to light the biologically variable underlying causes of widened sutures and persistent fontanelles leading to a common outcome. The authors herein present four syndromes, selected from a literature review, and their identified biological mechanisms in the context of altered suture physiology, exploring the roles of progenitor cell differentiation, extracellular matrix production, mineralization, and bone resorption. This article illustrates the gaps in understanding of complex craniofacial disorders, and the potential for further unification of genetics, cellular biology, and clinical pillars of health science research to improve treatment outcomes for patients.
Collapse
Affiliation(s)
- Daniela M. Roth
- School of Dentistry, University of Alberta, Edmonton, Canada
| | - Jeremie Oliver Piña
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | | | - Curtis Budden
- Department of Surgery, University of Alberta, Edmonton, Canada
| | - Daniel Graf
- School of Dentistry, University of Alberta, Edmonton, Canada
- Department of Medical Genetics, University of Alberta, Edmonton, Canada
| |
Collapse
|
6
|
Lee H, Hwa S, Cho S, Kim JH, Song HJ, Ko Y, Park JB. Impact of Polydeoxyribonucleotides on the Morphology, Viability, and Osteogenic Differentiation of Gingiva-Derived Stem Cell Spheroids. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1610. [PMID: 39459397 PMCID: PMC11509416 DOI: 10.3390/medicina60101610] [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: 09/14/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: Polydeoxyribonucleotides (PDRN), composed of DNA fragments derived from salmon DNA, is widely recognized for its regenerative properties. It has been extensively used in medical applications, such as dermatology and wound healing, due to its ability to enhance cellular metabolic activity, stimulate angiogenesis, and promote tissue regeneration. In the field of dentistry, PDRN has shown potential in promoting periodontal healing and bone regeneration. This study aims to investigate the effects of PDRN on the morphology, survival, and osteogenic differentiation of gingiva-derived stem cell spheroids, with a focus on its potential applications in tissue engineering and regenerative dentistry. Materials and Methods: Gingiva-derived mesenchymal stem cells were cultured and formed into spheroids using microwells. The cells were treated with varying concentrations of PDRN (0, 25, 50, 75, and 100 μg/mL) and cultivated in osteogenic media. Cell morphology was observed over seven days using an inverted microscope, and viability was assessed with Live/Dead Kit assays and Cell Counting Kit-8. Osteogenic differentiation was evaluated by measuring alkaline phosphatase activity and calcium deposition. The expression levels of osteogenic markers RUNX2 and COL1A1 were quantified using real-time polymerase chain reaction. RNA sequencing was performed to assess the gene expression profiles related to osteogenesis. Results: The results demonstrated that PDRN treatment had no significant effect on spheroid diameter or cellular viability during the observation period. However, a PDRN concentration of 75 μg/mL significantly enhanced calcium deposition by Day 14, suggesting increased mineralization. RUNX2 and COL1A1 mRNA expression levels varied with PDRN concentration, with the highest RUNX2 expression observed at 25 μg/mL and the highest COL1A1 expression at 75 μg/mL. RNA sequencing further confirmed the upregulation of genes involved in osteogenic differentiation, with enhanced expression of RUNX2 and COL1A1 in PDRN-treated gingiva-derived stem cell spheroids. Conclusions: In summary, PDRN did not significantly affect the viability or morphology of gingiva-derived stem cell spheroids but influenced their osteogenic differentiation and mineralization in a concentration-dependent manner. These findings suggest that PDRN may play a role in promoting osteogenic processes in tissue engineering and regenerative dentistry applications, with specific effects observed at different concentrations.
Collapse
Affiliation(s)
- Heera Lee
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.L.); (S.H.); (S.C.); (J.-H.K.); (Y.K.)
- Department of Medicine, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Somyeong Hwa
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.L.); (S.H.); (S.C.); (J.-H.K.); (Y.K.)
- Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sunga Cho
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.L.); (S.H.); (S.C.); (J.-H.K.); (Y.K.)
| | - Ju-Hwan Kim
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.L.); (S.H.); (S.C.); (J.-H.K.); (Y.K.)
| | - Hye-Jung Song
- Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Youngkyung Ko
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.L.); (S.H.); (S.C.); (J.-H.K.); (Y.K.)
- Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.L.); (S.H.); (S.C.); (J.-H.K.); (Y.K.)
- Department of Medicine, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
7
|
Tan YL, Ju SH, Wang Q, Zhong R, Gao JH, Wang MJ, Kang YL, Xu MZ. Shuanglongjiegu pill promoted bone marrow mesenchymal stem cell osteogenic differentiation by regulating the miR-217/RUNX2 axis to activate Wnt/β-catenin pathway. J Orthop Surg Res 2024; 19:617. [PMID: 39350234 PMCID: PMC11443779 DOI: 10.1186/s13018-024-05085-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
This study aimed to investigate the effects of Shuanglongjiegu pill (SLJGP) on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and explore its mechanism based on miR-217/RUNX2 axis. Results found that drug-containing serum of SLJGP promoted BMSCs viability with a dose-dependent effect. Under osteogenic differentiation conditions, SLJGP promoted the expression of ALP, OPN, BMP2, RUNX2, and the osteogenic differentiation ability of BMSCs. In addition, SLJGP significantly reduced miR-217 expression, and miR-217 directly targeted RUNX2. After treatment with miR-217 mimic, the promoting effects of SLJGP on proliferation and osteogenic differentiation of BMSCs were significantly inhibited. MiR-217 mimic co-treated with pcDNA-RUNX2 further confirmed that the miR-217/RUNX2 axis was involved in SLJGP to promote osteogenic differentiation of BMSCs. In addition, analysis of Wnt/β-catenin pathway protein expression showed that SLJGP activated the Wnt/β-catenin pathway through miR-217/RUNX2. In conclusion, SLJGP promoted osteogenic differentiation of BMSCs by regulating miR-217/RUNX2 axis and activating Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- You-Li Tan
- Department of Pharmacy, Affiliated Sport Hospital of CDSU, Chengdu Sport University, Chengdu, 610041, China.
| | - Shao-Hua Ju
- Department of Pharmacy, Affiliated Sport Hospital of CDSU, Chengdu Sport University, Chengdu, 610041, China
| | - Qiang Wang
- Department of Rehabilitation of sports medicine, Affiliated Sport Hospital of CDSU, Chengdu Sport University, Chengdu, 610041, China
| | - Rui Zhong
- Department of Orthopedics, Affiliated Sports Hospital of Chengdu Sport University, Chengdu, 610041, China
| | - Ji-Hai Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ming-Jian Wang
- Department of Pharmacy, Affiliated Sport Hospital of CDSU, Chengdu Sport University, Chengdu, 610041, China
| | - Ya-Lan Kang
- Department of Pharmacy, Affiliated Sport Hospital of CDSU, Chengdu Sport University, Chengdu, 610041, China
| | - Meng-Zhang Xu
- Department of Neck, Shoulder, Waist, and Leg Pain, Sichuan Province Orthopedic Hospital, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Platko K, Gyulay G, Lebeau PF, MacDonald ME, Lynn EG, Byun JH, Igdoura SA, Holden RM, Roubtsova A, Seidah NG, Krepinsky JC, Austin RC. GDF10 is a negative regulator of vascular calcification. J Biol Chem 2024; 300:107805. [PMID: 39307303 PMCID: PMC11541827 DOI: 10.1016/j.jbc.2024.107805] [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: 04/03/2024] [Revised: 08/23/2024] [Accepted: 09/11/2024] [Indexed: 10/27/2024] Open
Abstract
Cardiovascular mortality is particularly high and increasing in patients with chronic kidney disease, with vascular calcification (VC) as a major pathophysiologic feature. VC is a highly regulated biological process similar to bone formation involving osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs). We have previously demonstrated that loss of T-cell death-associated gene 51 (TDAG51) expression leads to an attenuation of medial VC. We now show a significant induction of circulating levels of growth differentiation factor 10 (GDF10) in TDAG51-/- mice, which was of interest due to its established role as an inhibitor of osteoblast differentiation. The objective of this study was to examine the role of GDF10 in the osteogenic transdifferentiation of VSMCs. Using primary mouse and human VSMCs, as well as ex vivo aortic ring cultures, we demonstrated that treatment with recombinant human (rh) GDF10 mitigated phosphate-mediated hydroxyapatite (HA) mineral deposition. Furthermore, ex vivo aortic rings from GDF10-/- mice exhibited increased HA deposition compared to C57BL/6J controls. To explain our observations, we identified that rhGDF10 treatment reduced protein expression of runt-related transcription factor 2, a key driver of osteogenic transdifferentiation of VSMCs and VC. In support of these findings, in vivo treatment with rhGDF10 attenuated VD3-induced VC. Furthermore, we demonstrated an increase in circulating GDF10 in patients with chronic kidney disease with clinically defined severe VC, as assessed by coronary artery calcium score. Thus, our studies identify GDF10 as a novel inhibitor of mineral deposition and as such, may represent a potential novel biomarker and therapeutic target for the detection and management of VC.
Collapse
Affiliation(s)
- Khrystyna Platko
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Gabriel Gyulay
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Paul F Lebeau
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Melissa E MacDonald
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Edward G Lynn
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Jae Hyun Byun
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Suleiman A Igdoura
- Department of Biology, McMaster University Medical Centre, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Rachel M Holden
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Anna Roubtsova
- The Institut de Recherches Cliniques de Montréal (IRCM), Affiliated with Université de Montréal, Montréal, Quebec, Canada
| | - Nabil G Seidah
- The Institut de Recherches Cliniques de Montréal (IRCM), Affiliated with Université de Montréal, Montréal, Quebec, Canada
| | - Joan C Krepinsky
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada.
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada.
| |
Collapse
|
9
|
Tollabi M, Poursalehi Z, Mehrafshar P, Bakhtiari R, Sarmadi VH, Tayebi L, Haramshahi SMA. Insight into the role of integrins and integrins-targeting biomaterials in bone regeneration. Connect Tissue Res 2024; 65:343-363. [PMID: 39297793 PMCID: PMC11541888 DOI: 10.1080/03008207.2024.2396002] [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: 04/16/2024] [Revised: 07/06/2024] [Accepted: 08/19/2024] [Indexed: 10/17/2024]
Abstract
Features of the extracellular matrix, along with biochemical factors, have a momentous impress in making genes on and/or off. The interaction of cells and the extracellular matrix is mediated by integrins. Therefore, these molecules have pivotal roles in regulating cell behaviors. Integrins include a group of molecules with a variety of characteristics that can affect different molecular cascades. Considering the importance of these molecules in tissue regeneration after injury, it is necessary to know well the integrins involved in the process of connecting cells to the extracellular matrix in each tissue.With the increase in life expectancy, bone tissue engineering has received more attention from researchers. Integrins are critical components in osteoblast differentiation, survival, and bone mechanotransduction. During osteogenic differentiation in stem cells, specific integrins facilitate multiple signaling pathways through their cytoplasmic domain, leading to the induction of osteogenic differentiation. Also, due to the importance of using biomaterials in bone tissue engineering, efforts have been made to design and use biomaterials with maximum interaction with integrins. Notably, the use of RGD peptide or fibronectin for surface modification is a well-established and commonly employed approach to manipulate integrin activity.This review article looks into integrins' role in bone development and regeneration. It then goes on to explore the complex mechanisms by which integrins contribute to these processes. In addition, this review discusses the use of natural and synthetic biomaterials that target integrins to promote bone regeneration.
Collapse
Affiliation(s)
- Mohammad Tollabi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Poursalehi
- Department of Applied Cell Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Parichehr Mehrafshar
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Vahid Hosseinpour Sarmadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Seyed Mohammad Amin Haramshahi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Hazrate Fatemeh Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Tian K, He X, Lin X, Chen X, Su Y, Lu Z, Chen Z, Zhang L, Li P, Ma L, Lan Z, Zhao X, Fen G, Hai Q, Xue D, Jin Q. Unveiling the Role of Sik1 in Osteoblast Differentiation: Implications for Osteoarthritis. Mol Cell Biol 2024; 44:411-428. [PMID: 39169784 PMCID: PMC11485870 DOI: 10.1080/10985549.2024.2385633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease characterized by subchondral osteosclerosis, mainly due to osteoblast activity. This research investigates the function of Sik1, a member of the AMP-activated protein kinase family, in OA. Proteomic analysis was conducted on clinical samples from 30 OA patients, revealing a negative correlation between Sik1 expression and OA. In vitro experiments utilized BMSCs to examine the effect of Sik1 on osteogenic differentiation. BMSCs were cultured and induced toward osteogenesis with specific media. Sik1 overexpression was achieved through lentiviral transfection, followed by analysis of osteogenesis-associated proteins using Western blotting, RT-qPCR, and alkaline phosphate staining. In vivo experiments involved destabilizing the medial meniscus in mice to establish an OA model, assessing the therapeutic potential of Sik1. The CT scans and histological staining were used to analyze subchondral bone alterations and cartilage damage. The findings show that Sik1 downregulation correlates with advanced OA and heightened osteogenic differentiation in BMSCs. Sik1 overexpression inhibits osteogenesis-related markers in vitro and reduces cartilage damage and subchondral osteosclerosis in vivo. Mechanistically, Sik1 modulates osteogenesis and subchondral bone changes through Runx2 activity regulation. The research emphasizes Sik1 as a promising target for treating OA, suggesting its involvement in controlling bone formation and changes in the subchondral osteosclerosis.
Collapse
Affiliation(s)
- Kuanmin Tian
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xiaoxin He
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xue Lin
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xiaolei Chen
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yajing Su
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Zhidong Lu
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Zhirong Chen
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Liang Zhang
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Peng Li
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Long Ma
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Zhibin Lan
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xin Zhao
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Gangning Fen
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qinqin Hai
- The Third Ward of Orthopaedic Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Di Xue
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qunhua Jin
- Institute of Osteoarthropathy, Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
- First Clinical Medical School, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| |
Collapse
|
11
|
Wanionok NE, Morel GR, Fernández JM. Osteoporosis and Alzheimer´s disease (or Alzheimer´s disease and Osteoporosis). Ageing Res Rev 2024; 99:102408. [PMID: 38969142 DOI: 10.1016/j.arr.2024.102408] [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: 02/28/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Alzheimer's disease (AD) and osteoporosis are two diseases that mainly affect elderly people, with increases in the occurrence of cases due to a longer life expectancy. Several epidemiological studies have shown a reciprocal association between both diseases, finding an increase in incidence of osteoporosis in patients with AD, and a higher burden of AD in osteoporotic patients. This epidemiological relationship has motivated the search for molecules, genes, signaling pathways and mechanisms that are related to both pathologies. The mechanisms found in these studies can serve to improve treatments and establish better patient care protocols.
Collapse
Affiliation(s)
- Nahuel E Wanionok
- Laboratorio de Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Cs. Exactas. Universidad Nacional de La Plata UNLP-CIC, Argentina
| | - Gustavo R Morel
- Biochemistry Research Institute of La Plata "Professor Doctor Rodolfo R. Brenner" (INIBIOLP), Argentina
| | - Juan M Fernández
- Laboratorio de Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Cs. Exactas. Universidad Nacional de La Plata UNLP-CIC, Argentina.
| |
Collapse
|
12
|
Kudiyarasu S, Karuppan Perumal MK, Rajan Renuka R, Manickam Natrajan P. Chitosan composite with mesenchymal stem cells: Properties, mechanism, and its application in bone regeneration. Int J Biol Macromol 2024; 275:133502. [PMID: 38960259 DOI: 10.1016/j.ijbiomac.2024.133502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/07/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Bone defects resulting from trauma, illness or congenital abnormalities represent a significant challenge to global health. Conventional treatments such as autographs and allografts have limitations, leading to the exploration of bone tissue engineering (BTE) as an alternative approach. This review aims to provide a comprehensive analysis of bone regeneration mechanisms with a focus on the role of chitosan-based biomaterials and mesenchymal stem cells (MSCs) in BTE. In addition, the physiochemical and biological properties of chitosan, its potential for bone regeneration when combined with other materials and the mechanisms through which MSCs facilitate bone regeneration were investigated. In addition, different methods of scaffold development and the incorporation of MSCs into chitosan-based scaffolds were examined. Chitosan has remarkable biocompatibility, biodegradability and osteoconductivity, making it an attractive choice for BTE. Interactions between transcription factors such as Runx2 and Osterix and signaling pathways such as the BMP and Wnt pathways regulate the differentiation of MSCs and bone regeneration. Various forms of scaffolding, including porous and fibrous injections, have shown promise in BTE. The synergistic combination of chitosan and MSCs in BTE has significant potential for addressing bone defects and promoting bone regeneration, highlighting the promising future of clinical challenges posed by bone defects.
Collapse
Affiliation(s)
- Sushmitha Kudiyarasu
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, 173, Agaram Road, Selaiyur, Chennai 600073, Tamil Nadu, India
| | - Manoj Kumar Karuppan Perumal
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - Remya Rajan Renuka
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India.
| | - Prabhu Manickam Natrajan
- Department of Clinical Sciences, College of Dentistry, Centre of Medical and Bio-allied Health Sciences and Research, Ajman University, Ajman, United Arab Emirates..
| |
Collapse
|
13
|
Ureiro-Cueto G, Rodil SE, Silva-Bermúdez P, Santana-Vázquez M, Hoz-Rodríguez L, Arzate H, Montoya-Ayala G. Amorphous titanium oxide (aTiO 2) thin films biofunctionalized with CAP-p15 induce mineralized-like differentiation of human oral mucosal stem cells (hOMSCs). Biomed Mater 2024; 19:055003. [PMID: 38917837 DOI: 10.1088/1748-605x/ad5bab] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/25/2024] [Indexed: 06/27/2024]
Abstract
Insufficient osseointegration of titanium-based implants is a factor conditioning their long-term success. Therefore, different surface modifications, such as multifunctional oxide coatings, calcium phosphates, and the addition of molecules such as peptides, have been developed to improve the bioactivity of titanium-based biomaterials. In this work, we investigate the behavior of human oral mucosal stem cells (hOMSCs) cultured on amorphous titanium oxide (aTiO2), surfaces designed to simulate titanium (Ti) surfaces, biofunctionalized with a novel sequence derived from cementum attachment protein (CAP-p15), exploring its impact on guiding hOMSCs towards an osteogenic phenotype. We carried out cell attachment and viability assays. Next, hOMSCs differentiation was assessed by red alizarin stain, ALP activity, and western blot analysis by evaluating the expression of RUNX2, BSP, BMP2, and OCN at the protein level. Our results showed that functionalized surfaces with CAP-p15 (1 µg ml-1) displayed a synergistic effect increasing cell proliferation and cell attachment, ALP activity, and expression of osteogenic-related markers. These data demonstrate that CAP-p15 and its interaction with aTiO2surfaces promote osteoblastic differentiation and enhanced mineralization of hOMSCs when compared to pristine samples. Therefore, CAP-p15 shows the potential to be used as a therapeutical molecule capable of inducing mineralized tissue regeneration onto titanium-based implants.
Collapse
Affiliation(s)
- Guadalupe Ureiro-Cueto
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Mexico
| | - Sandra E Rodil
- Instituto de Investigaciones en Materiales, UNAM, Mexico
| | - Phaedra Silva-Bermúdez
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico
| | - Maricela Santana-Vázquez
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Mexico
| | - Lia Hoz-Rodríguez
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Mexico
| | - Higinio Arzate
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Mexico
| | - Gonzalo Montoya-Ayala
- Laboratorio de Biología Periodontal y Tejidos Mineralizados, Facultad de Odontología, UNAM, Mexico
| |
Collapse
|
14
|
Janmohammadi M, Doostmohammadi N, Bahraminasab M, Nourbakhsh MS, Arab S, Asgharzade S, Ghanbari A, Satari A. Evaluation of new bone formation in critical-sized rat calvarial defect using 3D printed polycaprolactone/tragacanth gum-bioactive glass composite scaffolds. Int J Biol Macromol 2024; 270:132361. [PMID: 38750857 DOI: 10.1016/j.ijbiomac.2024.132361] [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: 01/09/2024] [Revised: 03/13/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024]
Abstract
Critical-sized bone defects are a major challenge in reconstructive bone surgery and usually fail to be treated due to limited remaining bone quality and extensive healing time. The combination of 3D-printed scaffolds and bioactive materials is a promising approach for bone tissue regeneration. In this study, 3D-printed alkaline-treated polycaprolactone scaffolds (M-PCL) were fabricated and integrated with tragacanth gum- 45S5 bioactive glass (TG-BG) to treat critical-sized calvarial bone defects in female adult Wistar rats. After a healing period of four and eight weeks, the new bone of blank, M-PCL, and M-PCL/TG-BG groups were harvested and assessed. Micro-computed tomography, histological, biochemical, and biomechanical analyses, gene expression, and bone matrix formation were used to assess bone regeneration. The micro-computed tomography results showed that the M-PCL/TG-BG scaffolds not only induced bone tissue formation within the bone defect but also increased BMD and BV/TV compared to blank and M-PCL groups. According to the histological analysis, there was no evidence of bony union in the calvarial defect regions of blank groups, while in M-PCL/TG-BG groups bony integration and repair were observed. The M-PCL/TG-BG scaffolds promoted the Runx2 and collagen type I expression as compared with blank and M-PCL groups. Besides, the bone regeneration in M-PCL/TG-BG groups correlated with TG-BG incorporation. Moreover, the use of M-PCL/TG-BG scaffolds promoted the biomechanical properties in the bone remodeling process. These data demonstrated that the M-PCL/TG-BG scaffolds serve as a highly promising platform for the development of bone grafts, supporting bone regeneration with bone matrix formation, and osteogenic features. Our results exhibited that the 3D-printed M-PCL/TG-BG scaffolds are a promising strategy for successful bone regeneration.
Collapse
Affiliation(s)
- Mahsa Janmohammadi
- Department of Biomedical Engineering, Faculty of New Sciences and Technologies, Semnan University, Semnan, Iran
| | - Nesa Doostmohammadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Marjan Bahraminasab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran; Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| | | | - Samaneh Arab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Samira Asgharzade
- Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; Department of Molecular Medicine, of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ali Ghanbari
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Atefeh Satari
- Department of Molecular Medicine, of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
15
|
Feng J, Zhang Q, Pu F, Zhu Z, Lu K, Lu WW, Tong L, Yu H, Chen D. Signalling interaction between β-catenin and other signalling molecules during osteoarthritis development. Cell Prolif 2024; 57:e13600. [PMID: 38199244 PMCID: PMC11150147 DOI: 10.1111/cpr.13600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/29/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Osteoarthritis (OA) is the most prevalent disorder of synovial joint affecting multiple joints. In the past decade, we have witnessed conceptual switch of OA pathogenesis from a 'wear and tear' disease to a disease affecting entire joint. Extensive studies have been conducted to understand the underlying mechanisms of OA using genetic mouse models and ex vivo joint tissues derived from individuals with OA. These studies revealed that multiple signalling pathways are involved in OA development, including the canonical Wnt/β-catenin signalling and its interaction with other signalling pathways, such as transforming growth factor β (TGF-β), bone morphogenic protein (BMP), Indian Hedgehog (Ihh), nuclear factor κB (NF-κB), fibroblast growth factor (FGF), and Notch. The identification of signalling interaction and underlying mechanisms are currently underway and the specific molecule(s) and key signalling pathway(s) playing a decisive role in OA development need to be evaluated. This review will focus on recent progresses in understanding of the critical role of Wnt/β-catenin signalling in OA pathogenesis and interaction of β-catenin with other pathways, such as TGF-β, BMP, Notch, Ihh, NF-κB, and FGF. Understanding of these novel insights into the interaction of β-catenin with other pathways and its integration into a complex gene regulatory network during OA development will help us identify the key signalling pathway of OA pathogenesis leading to the discovery of novel therapeutic strategies for OA intervention.
Collapse
Affiliation(s)
- Jing Feng
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of WuhanTongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Department of OrthopedicsWuhan No. 1 HospitalWuhanHubeiChina
| | - Qing Zhang
- Department of EmergencyRenmin Hospital, Wuhan UniversityWuhanHubeiChina
| | - Feifei Pu
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of WuhanTongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Department of OrthopedicsWuhan No. 1 HospitalWuhanHubeiChina
| | - Zhenglin Zhu
- Department of Orthopedic Surgerythe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Ke Lu
- Faculty of Pharmaceutical SciencesShenzhen Institute of Advanced TechnologyShenzhenChina
- Research Center for Computer‐aided Drug DiscoveryShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - William W. Lu
- Faculty of Pharmaceutical SciencesShenzhen Institute of Advanced TechnologyShenzhenChina
| | - Liping Tong
- Research Center for Computer‐aided Drug DiscoveryShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Huan Yu
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of WuhanTongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Department of OrthopedicsWuhan No. 1 HospitalWuhanHubeiChina
| | - Di Chen
- Faculty of Pharmaceutical SciencesShenzhen Institute of Advanced TechnologyShenzhenChina
- Research Center for Computer‐aided Drug DiscoveryShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| |
Collapse
|
16
|
Yu Y, Li X, Zheng M, Zhou L, Zhang J, Wang J, Sun B. The potential benefits and mechanisms of protein nutritional intervention on bone health improvement. Crit Rev Food Sci Nutr 2024; 64:6380-6394. [PMID: 36655469 DOI: 10.1080/10408398.2023.2168250] [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] [Indexed: 01/20/2023]
Abstract
Osteoporosis commonly occurs in the older people and severe patients, with the main reason of the imbalance of bone metabolism (the rate of bone resorption exceeding the rate of bone formation), resulting in a decrease in bone mineral density and destruction of bone microstructure and further leading to the increased risk of fragility fracture. Recent studies indicate that protein nutritional support is beneficial for attenuating osteoporosis and improving bone health. This review summarized the classical mechanisms of protein intervention for alleviating osteoporosis on both suppressing bone resorption and regulating bone formation related pathways (promoting osteoblasts generation and proliferation, enhancing calcium absorption, and increasing collagen and mineral deposition), as well as the potential novel mechanisms via activating autophagy of osteoblasts, altering bone related miRNA profiles, regulating muscle-bone axis, and modulating gut microbiota abundance. Protein nutritional intervention is expected to provide novel approaches for the prevention and adjuvant therapy of osteoporosis.
Collapse
Affiliation(s)
- Yonghui Yu
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing, China
| | - Xinping Li
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing, China
| | - Mengjun Zheng
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing, China
| | - Linyue Zhou
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing, China
| | - Jingjie Zhang
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing, China
| | - Jing Wang
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing, China
| | - Baoguo Sun
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
17
|
Chiticaru EA, Ioniță M. A Novel Approach Using Reduced Graphene Oxide for the Detection of ALP and RUNX2 Osteogenic Biomarkers. Curr Issues Mol Biol 2024; 46:4489-4505. [PMID: 38785540 PMCID: PMC11119758 DOI: 10.3390/cimb46050272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/30/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024] Open
Abstract
In this work, we propose a new technique involving the modification of commercial screen-printed carbon electrodes with electrochemically reduced graphene oxide to serve as the starting point of a future electrochemical biosensor for the detection of two osteogenic biomarkers: alkaline phosphatase (ALP) and Runt-related transcription factor 2 (RUNX2). The electrodes were characterized after each modification by cyclic voltammetry and electrochemical impedance spectroscopy, showing the appropriate electrochemical characteristics for each modification type. The results obtained from scanning electron microscopy, Raman spectroscopy, X-ray photoelectron spectroscopy, and contact angle measurements are well correlated with each other, demonstrating the successful modification of the electrodes with graphene oxide and its subsequent reduction. The bioreceptors were immobilized on the electrodes by physical adsorption, which was confirmed by electrochemical methods, structural characterization, and contact angle measurements. Finally, the functionalized electrodes were incubated with the specific target analytes and the detection relied on monitoring the electrochemical changes occurring after the hybridization process. Our results indicated that the pilot platform has the ability to detect the two biomarkers up to 1 nM, with increased sensitivity observed for RUNX2, suggesting that after further optimizations, it has a high potential to be employed as a future biosensor.
Collapse
Affiliation(s)
- Elena Alina Chiticaru
- Faculty of Medical Engineering, National University of Science and Technology Politehnica Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania;
| | - Mariana Ioniță
- Faculty of Medical Engineering, National University of Science and Technology Politehnica Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania;
- Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, Gh. Polizu 1-7, 011061 Bucharest, Romania
| |
Collapse
|
18
|
Aparicio-Bautista DI, Jiménez-Ortega RF, Becerra-Cervera A, Aquino-Gálvez A, de León-Suárez VP, Casas-Ávila L, Salmerón J, Hidalgo-Bravo A, Rivera-Paredez B, Velázquez-Cruz R. Interaction between MARK3 (rs11623869), PLCB4 (rs6086746) and GEMIN2 (rs2277458) variants with bone mineral density and serum 25-hidroxivitamin D levels in Mexican Mestizo women. Front Endocrinol (Lausanne) 2024; 15:1392063. [PMID: 38715801 PMCID: PMC11074919 DOI: 10.3389/fendo.2024.1392063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/03/2024] [Indexed: 06/04/2024] Open
Abstract
Introduction Understanding the genetic factors contributing to variations in bone mineral density (BMD) and vitamin D could provide valuable insights into the pathogenesis of osteoporosis. This study aimed to evaluate the association of single nucleotide variants in MARK3 (rs11623869), PLCB4 (rs6086746), and GEMIN2 (rs2277458) with BMD in Mexican women. Methods The gene-gene interaction was evaluated in these variants in serum 25(OH)D levels and BMD. A genetic risk score (GRS) was created on the basis of the three genetic variants. Genotyping was performed using predesigned TaqMan assays. Results A significant association was found between the rs6086746-A variant and BMD at the total hip, femoral neck, and lumbar spine, in women aged 45 years or older. However, no association was observed between the variants rs11623869 and rs2277458. The rs11623869 × rs2277458 interaction was associated with total hip (p=0.002) and femoral neck BMD (p=0.013). Similarly, for vitamin D levels, we observed an interaction between the variants rs6086746 × rs2277458 (p=0.021). GRS revealed a significant association with total hip BMD (p trend=0.003) and femoral neck BMD (p trend=0.006), as well as increased vitamin D levels (p trend=0.0003). These findings provide evidence of the individual and joint effect of the MARK3, PLCB4, and GEMIN2 variants on BMD and serum vitamin D levels in Mexican women. Discussion This knowledge could help to elucidate the interaction mechanism between BMD-related genetic variants and 25OHD, contributing to the determination of the pathogenesis of osteoporosis and its potential implications during early interventions.
Collapse
Affiliation(s)
- Diana I. Aparicio-Bautista
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Rogelio F. Jiménez-Ortega
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Departamento de Ciencias de la Acupuntura. Universidad Estatal del Valle de Ecatepec. Ecatepec de Morelos, Estado de Mexico, Mexico
| | - Adriana Becerra-Cervera
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Mexico City, Mexico
| | - Arnoldo Aquino-Gálvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | | | - Leonora Casas-Ávila
- Departamento de Medicina Genómica, Instituto Nacional de Rehabilitación, Mexico City, Mexico
| | - Jorge Salmerón
- Centro de Investigación en Políticas, Población y Salud, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alberto Hidalgo-Bravo
- Departamento de Medicina Genómica, Instituto Nacional de Rehabilitación, Mexico City, Mexico
| | - Berenice Rivera-Paredez
- Centro de Investigación en Políticas, Población y Salud, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| |
Collapse
|
19
|
Li Z, Kegui H, Piao W, Xuejiu W, Lim KT, Jin H. PAI-1 transfected-conditioned media promotes osteogenic differentiation of hBMSCs. Cell Biol Int 2024. [PMID: 38654436 DOI: 10.1002/cbin.12166] [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: 07/27/2023] [Revised: 02/28/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
Reconstruction of injured bone remains challenging in the clinic owing to the lack of suitable bone grafts. The utilization of PAI-1 transfected-conditioned media (P-CM) has demonstrated its ability to facilitate the differentiation process of mesenchymal stem cells (MSCs), potentially serving as a crucial mediator in tissue regeneration. This research endeavored to explore the therapeutic potential of P-CM concerning the differentiation of human bone marrow mesenchymal stem cells (hBMSCs). To assess new bone formation, a rat calvaria critical defect model was employed, while in vitro experiments involved the use of the alizarin Red-S mineral induction test. In the rat calvaria critical defect model, P-CM treatment resulted in significan new bone formation. In vitro, P-CM treated hBMSCs displayed robust osteogenesis compared to the control group, as demonstrated by the mineral induction test using alizarin Red-S. P-CM with hydroxyapatite/β-tricalcium phosphate/fibrin gel treatment significantly exhibited new bone formation, and the expression of osteogenic associated markers was enhanced in the P-CM-treated group. In conclusion, results demonstrate that P-CM treatment significantly enhanced the osteogenic differantiation efficiency and new bone formation, thus could be used as an ideal therapeutic biomolecule for constructing bone-specific implants, especially for orthopedic and dental applications.
Collapse
Affiliation(s)
- Zhang Li
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Hou Kegui
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Wang Piao
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Wang Xuejiu
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, South Korea
| | - Hexiu Jin
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
20
|
Qiu Z, Lin X, Zou L, Fu W, Lv H. Effect of graphene oxide/ poly-L-lactic acid composite scaffold on the biological properties of human dental pulp stem cells. BMC Oral Health 2024; 24:413. [PMID: 38575940 PMCID: PMC10993485 DOI: 10.1186/s12903-024-04197-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Tissue engineering has attracted recent attention as a promising bone repair and reconstruction approach. Dental pulp stem cells (DPSCs) are pluripotent and can differentiate into bone cells with the correct environment and substrate. Therefore, suitable scaffold materials are essential for fabricating functional three-dimensional (3D) tissue and tissue regeneration. Composite scaffolds consisting of biodegradable polymers are very promising constructs. This study aims to verify the biological function of human DPSCs seeded onto composite scaffolds based on graphene oxide (GO) and poly-L-lactic acid (PLLA). METHODS The surface morphology was observed under scanning electron microscopy (SEM). Chemical composition was evaluated with Fourier transform infrared (FTIR) spectroscopy. The biocompatibility of GO/PLLA scaffolds was assessed using phalloidin staining of cytoskeletal actin filaments, live/dead staining, and a CCK-8 assay. The effect of GO/PLLA scaffolds on cell osteogenic differentiation was detected through ALP staining, ALP activity assays, and alizarin red S staining, complemented by quantitative real-time PCR (qRT-PCR) analysis. RESULTS Our data showed that GO and PLLA are successfully integrated and the GO/PLLA scaffolds exhibit favorable bioactivity and biocompatibility towards DPSCs. Additionally, it was observed that the 0.15% GO/PLLA scaffold group promoted DPSC proliferation and osteogenic differentiation by forming more calcium nodules, showing a higher intensity of ALP staining and ALP activity, and enhancing the expression levels of differentiation marker genes RUNX2 and COL1. CONCLUSIONS These results demonstrate that the GO/PLLA scaffold is a feasible composite material suitable for cell culture and holds promising applications for oral bone tissue engineering.
Collapse
Affiliation(s)
- Zailing Qiu
- Oral Center, Fujian Provincial Governmental Hospital, Fuzhou, China.
| | - Xuemei Lin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Luning Zou
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Weihao Fu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Hongbing Lv
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
21
|
Wu Y, Chen D, Li L. Morinda officinalis polysaccharide promotes the osteogenic differentiation of human bone marrow-derived mesenchymal stem cells via microRNA-210-3p/scavenger receptor class A member 3. J Investig Med 2024; 72:370-382. [PMID: 38264863 DOI: 10.1177/10815589241229693] [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] [Indexed: 01/25/2024]
Abstract
Morinda officinalis polysaccharide (MOP) is the bioactive ingredient extracted from the root of Morinda officinalis, and Morinda officinalis is applied to treat osteoporosis (OP). The purpose of this study was to determine the role of MOP on human bone marrow mesenchymal stem cells (hBMSCs) and the underlying mechanism. HBMSCs were isolated from bone marrow samples of patients with OP and treated with MOP. Quantitative real-time polymerase chain reaction was adopted to quantify the expression of microRNA-210-3p (miR-210-3p) and scavenger receptor class A member 3 (SCARA3) mRNA. Cell Counting Kit-8 assay was employed to detect cell viability; Terminal-deoxynucleotidyl Transferase Mediated Nick End Labeling assay and flow cytometry were adopted to detect apoptosis; Alkaline Phosphatase (ALP) activity assay kit was applied to detect ALP activity; Western blot was executed to quantify the expression levels of SCARA3, osteogenic and adipogenic differentiation markers. Ovariectomized rats were treated with MOP. Bone mineral density (BMD), serum tartrate-resistant acid phosphatase 5b (TRACP 5b), and N-telopeptide of type I collagen (NTx) levels were assessed by BMD detector and Enzyme-linked immunosorbent assay kits. It was revealed that MOP could promote hBMSCs' viability and osteogenic differentiation and inhibit apoptosis and adipogenic differentiation. MOP could also upregulate SCARA3 expression through repressing miR-210-3p expression. Treatment with MOP increased the BMD and decreased the TRACP 5b and NTx levels in ovariectomized rats. MOP may boost the osteogenic differentiation and inhibit adipogenic differentiation of hBMSCs by miR-210-3p/SCARA3 axis.
Collapse
Affiliation(s)
- Yue Wu
- Department of Thoracic Surgery, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Dan Chen
- Department of Rehabilitation, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Longguang Li
- Department of Rehabilitation, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
22
|
Tian Y, Cui Y, Ren G, Fan Y, Dou M, Li S, Wang G, Wang Y, Peng C, Wu D. Dual-functional thermosensitive hydrogel for reducing infection and enhancing bone regeneration in infected bone defects. Mater Today Bio 2024; 25:100972. [PMID: 38312799 PMCID: PMC10835005 DOI: 10.1016/j.mtbio.2024.100972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/19/2023] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
The contamination of bone defects is a serious therapeutic problem. The treatment of infected bone defects involves rigorous infection control followed by bone reconstruction. Considering these two processes, the development of biomaterials possessing antibacterial and osteogenic properties offers a promising approach for the treatment of infected bone defects. In this study, a dual-functional, thermosensitive, and injectable hydrogel composed of chitosan (CS), quaternized CS (QCS), and nano-hydroxyapatite (nHA) was designed, and the ratio of CS to QCS in the hydrogel was optimized to enhance the antibacterial efficacy of CS while reducing the cytotoxicity of QCS. In vitro studies demonstrated that the hydrogel with an 85 %:15 % ratio of CS to QCS exhibited excellent biocompatibility and antibacterial properties while also possessing suitable mechanical characteristics and degradability. The incorporation of nHA into the hydrogel enhanced MC3T3-E1 proliferation and osteogenic differentiation. Moreover, this hydrogel demonstrated superior in vivo therapeutic effectiveness in a rabbit model of infected bone defect. In summary, this study provides a promising material design and a comprehensive one-step treatment strategy for infected bone defects.
Collapse
Affiliation(s)
- Yuhang Tian
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Yutao Cui
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Guangkai Ren
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Yi Fan
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Minghan Dou
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Shaorong Li
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Gan Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Yanbing Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Chuangang Peng
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Dankai Wu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| |
Collapse
|
23
|
Toral López J, Gómez Martinez S, Rivera Vega MDR, Hernández-Zamora E, Cuevas Covarrubias S, Ibarra Castrejón BA, González Huerta LM. New Genetic Variants of RUNX2 in Mexican Families Cause Cleidocranial Dysplasia. BIOLOGY 2024; 13:173. [PMID: 38534443 DOI: 10.3390/biology13030173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024]
Abstract
Cleidocranial dysplasia (CCD) is an autosomal dominant skeletal dysplasia characterized by persistent open skull sutures with bulging calvaria, hypoplasia, or aplasia of clavicles permitting abnormal opposition of the shoulders; wide public symphysis; short middle phalanx of the fifth fingers; and vertebral, craniofacial, and dental anomalies. It is a rare disease, with a prevalence of 1-9/1,000,000, high penetrance, and variable expression. The gene responsible for CCD is the Runt-related transcription factor 2 (RUNX2) gene. We characterize the clinical, genetic, and bioinformatic results of four CCD cases: two cases within Mexican families with six affected members, nine asymptomatic individuals, and two sporadic cases with CCD, with one hundred healthy controls. Genomic DNA analyses of the RUNX2 gene were performed for Sanger sequencing. Bioinformatics tools were used to predict the function, stability, and structural changes of the mutated RUNX2 proteins. Three novel heterozygous mutations (c.651_652delTA; c.538_539delinsCA; c.662T>A) and a previously reported mutation (c.674G>A) were detected. In silico analysis showed that all mutations had functional, stability-related, and structural alterations in the RUNX2 protein. Our results show novel mutations that enrich the pool of RUNX2 gene mutations with CCD. Moreover, the proband 1 presented clinical data not previously reported that could represent an expanded phenotype of severe expression.
Collapse
Affiliation(s)
- Jaime Toral López
- Department of Medical Genetics, Centro Médico Ecatepec ISSEMYM, Ecatepec 55000, México State, Mexico
| | - Sandra Gómez Martinez
- Servicio de Genética, Hospital General de México "Eduardo Liceaga" (HGM), México City 06720, Mexico
| | | | - Edgar Hernández-Zamora
- Medicina Genómica, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", México City 14389, Mexico
| | | | | | | |
Collapse
|
24
|
Zhou S, Xiao C, Fan L, Yang J, Ge R, Cai M, Yuan K, Li C, Crawford RW, Xiao Y, Yu P, Deng C, Ning C, Zhou L, Wang Y. Injectable ultrasound-powered bone-adhesive nanocomposite hydrogel for electrically accelerated irregular bone defect healing. J Nanobiotechnology 2024; 22:54. [PMID: 38326903 PMCID: PMC10851493 DOI: 10.1186/s12951-024-02320-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/26/2024] [Indexed: 02/09/2024] Open
Abstract
The treatment of critical-size bone defects with irregular shapes remains a major challenge in the field of orthopedics. Bone implants with adaptability to complex morphological bone defects, bone-adhesive properties, and potent osteogenic capacity are necessary. Here, a shape-adaptive, highly bone-adhesive, and ultrasound-powered injectable nanocomposite hydrogel is developed via dynamic covalent crosslinking of amine-modified piezoelectric nanoparticles and biopolymer hydrogel networks for electrically accelerated bone healing. Depending on the inorganic-organic interaction between the amino-modified piezoelectric nanoparticles and the bio-adhesive hydrogel network, the bone adhesive strength of the prepared hydrogel exhibited an approximately 3-fold increase. In response to ultrasound radiation, the nanocomposite hydrogel could generate a controllable electrical output (-41.16 to 61.82 mV) to enhance the osteogenic effect in vitro and in vivo significantly. Rat critical-size calvarial defect repair validates accelerated bone healing. In addition, bioinformatics analysis reveals that the ultrasound-responsive nanocomposite hydrogel enhanced the osteogenic differentiation of bone mesenchymal stem cells by increasing calcium ion influx and up-regulating the PI3K/AKT and MEK/ERK signaling pathways. Overall, the present work reveals a novel wireless ultrasound-powered bone-adhesive nanocomposite hydrogel that broadens the therapeutic horizons for irregular bone defects.
Collapse
Affiliation(s)
- Shiqi Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China
| | - Cairong Xiao
- School of Materials Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510641, China
| | - Lei Fan
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jinghong Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China
| | - Ruihan Ge
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China
| | - Min Cai
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China
| | - Kaiting Yuan
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China
| | - Changhao Li
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China
| | - Ross William Crawford
- Institute of Health and Biomedical Innovation & Australia-China Centre for Tissue Engineering and Regenerative Medicine, Centre for Biomedical Technologies, Queensland University of Technology, Queensland, 4059, Australia
| | - Yin Xiao
- School of Medicine and Dentistry & Menzies Health Institute Queensland, Griffith University, Queensland, 4111, Australia
| | - Peng Yu
- School of Materials Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510641, China
| | - Chunlin Deng
- School of Materials Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510641, China
| | - Chengyun Ning
- School of Materials Science and Engineering, National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510641, China.
| | - Lei Zhou
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Spine Surgery, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510150, China.
| | - Yan Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China.
| |
Collapse
|
25
|
Li A, Hancock JC, Quezado M, Ahn S, Briceno N, Celiku O, Ranjan S, Aboud O, Colwell N, Kim SA, Nduom E, Kuhn S, Park DM, Vera E, Aldape K, Armstrong TS, Gilbert MR. TGF-β and BMP signaling are associated with the transformation of glioblastoma to gliosarcoma and then osteosarcoma. Neurooncol Adv 2024; 6:vdad164. [PMID: 38292240 PMCID: PMC10825841 DOI: 10.1093/noajnl/vdad164] [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] [Indexed: 02/01/2024] Open
Abstract
Background Gliosarcoma, an isocitrate dehydrogenase wildtype (IDH-WT) variant of glioblastoma, is defined by clonal biphasic differentiation into gliomatous and sarcomatous components. While the transformation from a glioblastoma to gliosarcoma is uncommon, the subsequent transformation to osteosarcoma is rare but may provide additional insights into the biology of these typically distinct cancers. We observed a patient initially diagnosed with glioblastoma, that differentiated into gliosarcoma at recurrence, and further evolved to osteosarcoma at the second relapse. Our objective was to characterize the molecular mechanisms of tumor progression associated with this phenotypic transformation. Methods Tumor samples were collected at all 3 stages of disease and RNA sequencing was performed to capture their transcriptomic profiles. Sequential clonal evolution was confirmed by the maintenance of an identical PTEN mutation throughout the tumor differentiation using the TSO500 gene panel. Publicly available datasets and the Nanostring nCounter technology were used to validate the results. Results The glioblastoma tumor from this patient possessed mixed features of all 3 TCGA-defined transcriptomic subtypes of an IDH-WT glioblastoma and a proportion of osteosarcoma signatures were upregulated in the original tumor. Analysis showed that enhanced transforming growth factor-β (TGF-β) and bone morphogenic protein signaling was associated with tumor transformation. Regulatory network analysis revealed that TGF-β family signaling committed the lineage tumor to osteogenesis by stimulating the expression of runt-related transcription factor 2 (RUNX2), a master regulator of bone formation. Conclusions This unusual clinical case provided an opportunity to explore the modulators of longitudinal sarcomatous transformation, potentially uncovering markers indicating predisposition to this change and identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Aiguo Li
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - John C Hancock
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Martha Quezado
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Susie Ahn
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Nicole Briceno
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Orieta Celiku
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Surabhi Ranjan
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Orwa Aboud
- Department of Neurology and Neurological Surgery, University of California, Davis, Sacramento, California, USA
| | - Nicole Colwell
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Sun A Kim
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Edjah Nduom
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Skyler Kuhn
- Research Technology Branch, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Deric M Park
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Elizabeth Vera
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Ken Aldape
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Terri S Armstrong
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
26
|
Bi M, Yang K, Yu T, Wu G, Li Q. Cell-based mechanisms and strategies of co-culture system both in vivo and vitro for bone tissue engineering. Biomed Pharmacother 2023; 169:115907. [PMID: 37984308 DOI: 10.1016/j.biopha.2023.115907] [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: 09/09/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023] Open
Abstract
The lack of a functional vascular supply has been identified as a major challenge limiting the clinical introduction of stem cell-based bone tissue engineering (BTE) for the repair of large-volume bone defects (LVBD). Various approaches have been explored to improve the vascular supply in tissue-engineered constructs, and the development of strategies that could effectively induce the establishment of a functional vascular supply has become a major goal of BTE research. One of the state-of-the-art methods is to incorporate both angiogenic and osteogenic cells in co-culture systems. This review clarifies the key concepts involved, summarises the cell types and models used to date, and systematically evaluates their performance. We also discuss the cell-to-cell communication between these two cell types and the strategies explored in BTE constructs with angiogenic and osteogenic cells to optimise their functions. In addition, we outline unresolved issues and remaining obstacles that need to be overcome for further development in this field and eventual successful repair of LVBD.
Collapse
Affiliation(s)
- Mengning Bi
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China; Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology Shanghai, China
| | - Kaiwen Yang
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China; Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai Key Laboratory of Stomatology &Shanghai Research Institute of Stomatology; National Clinical Research Center of Stomatology, Shanghai, China
| | - Tao Yu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands; Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam, the Netherlands.
| | - Qiong Li
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China.
| |
Collapse
|
27
|
Xiao Y, Xie X, Chen Z, Yin G, Kong W, Zhou J. Advances in the roles of ATF4 in osteoporosis. Biomed Pharmacother 2023; 169:115864. [PMID: 37948991 DOI: 10.1016/j.biopha.2023.115864] [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: 09/07/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023] Open
Abstract
Osteoporosis (OP) is characterized by reduced bone mass, decreased strength, and enhanced bone fragility fracture risk. Activating transcription factor 4 (ATF4) plays a role in cell differentiation, proliferation, apoptosis, redox balance, amino acid uptake, and glycolipid metabolism. ATF4 induces the differentiation of bone marrow mesenchymal stem cells (BM-MSCs) into osteoblasts, increases osteoblast activity, and inhibits osteoclast formation, promoting bone formation and remodeling. In addition, ATF4 mediates the energy metabolism in osteoblasts and promotes angiogenesis. ATF4 is also involved in the mediation of adipogenesis. ATF4 can selectively accumulate in osteoblasts. ATF4 can directly interact with RUNT-related transcription factor 2 (RUNX2) and up-regulate the expression of osteocalcin (OCN) and osterix (Osx). Several upstream factors, such as Wnt/β-catenin and BMP2/Smad signaling pathways, have been involved in ATF4-mediated osteoblast differentiation. ATF4 promotes osteoclastogenesis by mediating the receptor activator of nuclear factor κ-B (NF-κB) ligand (RANKL) signaling. Several agents, such as parathyroid (PTH), melatonin, and natural compounds, have been reported to regulate ATF4 expression and mediate bone metabolism. In this review, we comprehensively discuss the biological activities of ATF4 in maintaining bone homeostasis and inhibiting OP development. ATF4 has become a therapeutic target for OP treatment.
Collapse
Affiliation(s)
- Yaosheng Xiao
- Department of Orthopaetics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Xunlu Xie
- Department of Pathology, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Zhixi Chen
- Department of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Guoqiang Yin
- Ganzhou Hospital Affiliated to Nanchang University, Ganzhou 341000, China
| | - Weihao Kong
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| |
Collapse
|
28
|
Ding S, Ma Y, Yang J, Tang Y, Jin Y, Li L, Ma C. MiR-224-5p inhibits osteoblast differentiation and impairs bone formation by targeting Runx2 and Sp7. Cytotechnology 2023; 75:505-516. [PMID: 37841957 PMCID: PMC10575840 DOI: 10.1007/s10616-023-00593-z] [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: 08/17/2022] [Accepted: 08/24/2023] [Indexed: 10/17/2023] Open
Abstract
Osteoporosis is a complicated multifactorial disorder characterized by low bone mass and deteriorated bone microarchitecture with an elevated fracture risk. MicroRNAs play important roles in osteoblastic differentiation. In the present study, we found that miR-224-5p was markedly downregulated during the osteogenic differentiation of C2C12 cells. Overexpression of miR-224-5p in C2C12 cells inhibited osteoblast activity, as indicated by reduced ALP activity, matrix mineralization and the expression of osteogenic marker genes. Moreover, we demonstrated that Runx2 and Sp7 were direct targets of miR-224-5p. Furthermore, the specific inhibition of miR-224-5p by femoral bone marrow cavity injection with miR-224-5p antagomir prevented ovariectomy-induced bone loss. Finally, we found that the levels of miR-224-5p were markedly elevated in the sera of patients with osteoporosis. Collectively, this study revealed that miR-224-5p negatively regulates osteogenic differentiation by targeting Runx2 and Sp7. It also highlights the potential use of miR-224-5p as a therapeutic target and diagnostic biomarker for osteoporosis. Supplementary information The online version contains supplementary material available at 10.1007/s10616-023-00593-z.
Collapse
Affiliation(s)
- Siyang Ding
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
- Jiangsu Key Laboratory of Oral Disease, Department of Sixth Outpatient, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210029 China
| | - Yunfei Ma
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| | - Jiashu Yang
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| | - Yuting Tang
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| | - Yucui Jin
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| | - Lingyun Li
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| | - Changyan Ma
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| |
Collapse
|
29
|
Khotib J, Marhaeny HD, Miatmoko A, Budiatin AS, Ardianto C, Rahmadi M, Pratama YA, Tahir M. Differentiation of osteoblasts: the links between essential transcription factors. J Biomol Struct Dyn 2023; 41:10257-10276. [PMID: 36420663 DOI: 10.1080/07391102.2022.2148749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/12/2022] [Indexed: 11/27/2022]
Abstract
Osteoblasts, cells derived from mesenchymal stem cells (MSCs) in the bone marrow, are cells responsible for bone formation and remodeling. The differentiation of osteoblasts from MSCs is triggered by the expression of specific genes, which are subsequently controlled by pro-osteogenic pathways. Mature osteoblasts then differentiate into osteocytes and are embedded in the bone matrix. Dysregulation of osteoblast function can cause inadequate bone formation, which leads to the development of bone disease. Various key molecules are involved in the regulation of osteoblastogenesis, which are transcription factors. Previous studies have heavily examined the role of factors that control gene expression during osteoblastogenesis, both in vitro and in vivo. However, the systematic relationship of these transcription factors remains unknown. The involvement of ncRNAs in this mechanism, particularly miRNAs, lncRNAs, and circRNAs, has been shown to influence transcriptional factor activity in the regulation of osteoblast differentiation. Here, we discuss nine essential transcription factors involved in osteoblast differentiation, including Runx2, Osx, Dlx5, β-catenin, ATF4, Ihh, Satb2, and Shn3. In addition, we summarize the role of ncRNAs and their relationship to these essential transcription factors in order to improve our understanding of the transcriptional regulation of osteoblast differentiation. Adequate exploration and understanding of the molecular mechanisms of osteoblastogenesis can be a critical strategy in the development of therapies for bone-related diseases.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Junaidi Khotib
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Honey Dzikri Marhaeny
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Andang Miatmoko
- Department of Pharmaceutical Science, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Aniek Setiya Budiatin
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Mahardian Rahmadi
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Yusuf Alif Pratama
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Muhammad Tahir
- Department of Pharmaceutical Science, Kulliyah of Pharmacy, International Islamic University Malaysia, Pahang, Malaysia
| |
Collapse
|
30
|
Ghuloum FI, Stevens LA, Johnson CA, Riobo-Del Galdo NA, Amer MH. Towards modular engineering of cell signalling: Topographically-textured microparticles induce osteogenesis via activation of canonical hedgehog signalling. BIOMATERIALS ADVANCES 2023; 154:213652. [PMID: 37837904 DOI: 10.1016/j.bioadv.2023.213652] [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: 07/19/2023] [Revised: 09/19/2023] [Accepted: 10/03/2023] [Indexed: 10/16/2023]
Abstract
Polymer microparticles possess great potential as functional building blocks for advanced bottom-up engineering of complex tissues. Tailoring the three-dimensional architectural features of culture substrates has been shown to induce osteogenesis in mesenchymal stem cells in vitro, but the molecular mechanisms underpinning this remain unclear. This study proposes a mechanism linking the activation of Hedgehog signalling to the osteoinductive effect of surface-engineered, topographically-textured polymeric microparticles. In this study, mesenchymal progenitor C3H10T1/2 cells were cultured on smooth and dimpled poly(D,l-lactide) microparticles to assess differences in viability, cellular morphology, proliferation, and expression of a range of Hedgehog signalling components and osteogenesis-related genes. Dimpled microparticles induced osteogenesis and activated the Hedgehog signalling pathway relative to smooth microparticles and 2D-cultured controls without the addition of exogenous biochemical factors. We observed upregulation of the osteogenesis markers Runt-related transcription factor2 (Runx2) and bone gamma-carboxyglutamate protein 2 (Bglap2), as well as the Hedgehog signalling components, glioma associated oncogene homolog 1 (Gli1), Patched1 (Ptch1), and Smoothened (Smo). Treatment with the Smo antagonist KAAD-cyclopamine confirmed the involvement of Smo in Gli1 target gene activation, with a significant reduction in the expression of Gli1, Runx2 and Bglap2 (p ≤ 0.001) following KAAD-cyclopamine treatment. Overall, our study demonstrates the role of the topographical microenvironment in the modulation of Hedgehog signalling, highlighting the potential for tailoring substrate topographical design to offer cell-instructive 3D microenvironments. Topographically-textured microparticles allow the modulation of Hedgehog signalling in vitro without adding exogenous biochemical agonists, thereby eliminating potential confounding artefacts in high-throughput drug screening applications.
Collapse
Affiliation(s)
- Fatmah I Ghuloum
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom; Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Lee A Stevens
- Low Carbon Energy and Resources Technologies Research Group, Faculty of Engineering, University of Nottingham, UK
| | - Colin A Johnson
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Natalia A Riobo-Del Galdo
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom; Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, UK
| | - Mahetab H Amer
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
31
|
Roldan L, Montoya C, Solanki V, Cai KQ, Yang M, Correa S, Orrego S. A Novel Injectable Piezoelectric Hydrogel for Periodontal Disease Treatment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43441-43454. [PMID: 37672788 DOI: 10.1021/acsami.3c08336] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Periodontal disease is a multifactorial, bacterially induced inflammatory condition characterized by the progressive destruction of periodontal tissues. The successful nonsurgical treatment of periodontitis requires multifunctional technologies offering antibacterial therapies and promotion of bone regeneration simultaneously. For the first time, in this study, an injectable piezoelectric hydrogel (PiezoGEL) was developed after combining gelatin methacryloyl (GelMA) with biocompatible piezoelectric fillers of barium titanate (BTO) that produce electrical charges when stimulated by biomechanical vibrations (e.g., mastication, movements). We harnessed the benefits of hydrogels (injectable, light curable, conforms to pocket spaces, biocompatible) with the bioactive effects of piezoelectric charges. A thorough biomaterial characterization confirmed piezoelectric fillers' successful integration with the hydrogel, photopolymerizability, injectability for clinical use, and electrical charge generation to enable bioactive effects (antibacterial and bone tissue regeneration). PiezoGEL showed significant reductions in pathogenic biofilm biomass (∼41%), metabolic activity (∼75%), and the number of viable cells (∼2-3 log) compared to hydrogels without BTO fillers in vitro. Molecular analysis related the antibacterial effects to be associated with reduced cell adhesion (downregulation of porP and fimA) and increased oxidative stress (upregulation of oxyR) genes. Moreover, PiezoGEL significantly enhanced bone marrow stem cell (BMSC) viability and osteogenic differentiation by upregulating RUNX2, COL1A1, and ALP. In vivo, PiezoGEL effectively reduced periodontal inflammation and increased bone tissue regeneration compared to control groups in a mice model. Findings from this study suggest PiezoGEL to be a promising and novel therapeutic candidate for the treatment of periodontal disease nonsurgically.
Collapse
Affiliation(s)
- Lina Roldan
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
- Bioengineering Research Group (GIB), Universidad EAFIT, Medellín 050037, Colombia
| | - Carolina Montoya
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Varun Solanki
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Maobin Yang
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
- Department of Endodontology, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Santiago Correa
- Bioengineering Research Group (GIB), Universidad EAFIT, Medellín 050037, Colombia
| | - Santiago Orrego
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
- Bioengineering Department, College of Engineering, Temple University. Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
32
|
Abnosi MH, Sargolzaei J, Nazari F. Gallic Acid Ameliorates Cadmium Effect on Osteogenesis by Activation of Alkaline Phosphatase and Collagen Synthesis. CELL JOURNAL 2023; 25:603-612. [PMID: 37718763 PMCID: PMC10520984 DOI: 10.22074/cellj.2023.1999110.1263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/17/2023] [Accepted: 07/26/2023] [Indexed: 09/19/2023]
Abstract
OBJECTIVE We previously reported that cadmium (Cd) inhibits osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). In addition, gallic acid (GA) improves BMSC differentiation. Here, we aim to study the ability of GA to prevent osteogenic inhibition induced by Cd. MATERIALS AND METHODS In this experimental study, BMSCs were extracted and purified from Wistar rats and their viability was determined in the presence of Cd and GA. The results indicated that 1.5 μM Cd and 0.25 μM of GA were appropriate for further investigation. After 20 days in osteogenic medium, matrix production was analysed by alizarin red, calcium content, and alkaline phosphatase (ALP) activity. Osteogenic-related genes and collagen 1A1 (COL1A1) protein expressions were investigated. The preventive effect of GA on oxidative stress and metabolic change induced by Cd was estimated. RESULTS GA counteracted the inhibitory effect of Cd on matrix production and significantly (P=0.0001) improved the osteogenic differentiation ability of BMSCs. Also, GA prevented the toxic effect of Cd on osteogenic-related gene expressions and nullified the reducing effect of Cd on COL1Al and ALP activity. A significant reduction (P=0.0001) in malondialdehyde and lactic acid concentration showed that GA counteracted both oxidative stress and metabolic changes caused by Cd. CONCLUSION GA prevented the toxic effect of Cd, an environmental pollutant and a factor in osteoporosis.
Collapse
Affiliation(s)
| | - Javad Sargolzaei
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| | - Farshid Nazari
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| |
Collapse
|
33
|
Malavašič P, Polajžer S, Lovšin N. Anaphase-Promoting Complex Subunit 1 Associates with Bone Mineral Density in Human Osteoporotic Bone. Int J Mol Sci 2023; 24:12895. [PMID: 37629076 PMCID: PMC10454667 DOI: 10.3390/ijms241612895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Genome-wide association studies (GWAS) are one of the most common approaches to identify genetic loci that are associated with bone mineral density (BMD). Such novel genetic loci represent new potential targets for the prevention and treatment of fragility fractures. GWAS have identified hundreds of associations with BMD; however, only a few have been functionally evaluated. A locus significantly associated with femoral neck BMD at the genome-wide level is intronic SNP rs17040773 located in the intronic region of the anaphase-promoting complex subunit 1 (ANAPC1) gene (p = 1.5 × 10-9). Here, we functionally evaluate the role of ANAPC1 in bone remodelling by examining the expression of ANAPC1 in human bone and muscle tissues and during the osteogenic differentiation of human primary mesenchymal stem cells (MSCs). The expression of ANAPC1 was significantly decreased 2.3-fold in bone tissues and 6.2-fold in muscle tissue from osteoporotic patients as compared to the osteoarthritic and control tissues. Next, we show that the expression of ANAPC1 changes during the osteogenic differentiation process of human MSCs. Moreover, the silencing of ANAPC1 in human osteosarcoma (HOS) cells reduced RUNX2 expression, suggesting that ANAPC1 affects osteogenic differentiation through RUNX2. Altogether, our results indicate that ANAPC1 plays a role in bone physiology and in the development of osteoporosis.
Collapse
Affiliation(s)
- Petra Malavašič
- General Hospital Novo Mesto, Šmihelska Cesta 1, 8000 Novo Mesto, Slovenia;
| | - Sara Polajžer
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia
| | - Nika Lovšin
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
34
|
Guo S, Yan M, Li X, Zhang S, Liu Z, Li K, Liu P, Liu Y, Sun G, Fu Q. Single-cell RNA-seq analysis reveals that immune cells induce human nucleus pulposus ossification and degeneration. Front Immunol 2023; 14:1224627. [PMID: 37638033 PMCID: PMC10449260 DOI: 10.3389/fimmu.2023.1224627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Background and aims Determining the transcriptomes and molecular mechanism underlying human degenerative nucleus pulposus (NP) is of critical importance for treating intervertebral disc degeneration (IDD). Here, we aimed to elucidate the detailed molecular mechanism of NP ossification and IDD using single-cell RNA sequencing. Methods Single-cell RNA-seq and bioinformatic analysis were performed to identify NP cell populations with gene signatures, biological processes and pathways, and subpopulation analysis, RNA velocity analysis, and cell-to-cell communication analysis were performed in four IDD patients. We also verified the effects of immune cells on NP ossification using cultured NP cells and a well-established rat IDD model. Results We identified five cell populations with gene expression profiles in degenerative NP at single-cell resolution. GO database analysis showed that degenerative NP-associated genes were mainly enriched in extracellular matrix organization, immune response, and ossification. Gene set enrichment analysis showed that rheumatoid arthritis signaling, antigen processing and presentation signaling were activated in the blood cell cluster. We revealed that stromal cells, which are progenitor cells, differentiated toward an ossification phenotype and delineated interactions between immune cells (macrophages and T cells) and stromal cells. Immune factors such as TNF-α, CD74 and CCL-3 promoted the differentiation of stromal cells toward an ossification phenotype in vitro. Blocking TNF-α with a specific inhibitor successfully reversed NP ossification and modified NP morphology in vivo. Conclusion Our study revealed an increase in macrophages and T cells in degenerative NP, which induced stromal cell differentiation toward an ossification phenotype, and contributed to the identification of a novel therapeutic target to delay IDD.
Collapse
Affiliation(s)
- Song Guo
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Meijun Yan
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Xinhua Li
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Shuya Zhang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Zhong Liu
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Kewei Li
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Pengcheng Liu
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Yanbin Liu
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Guixin Sun
- Department of Traumatology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiang Fu
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| |
Collapse
|
35
|
Thaweesapphithak S, Theerapanon T, Rattanapornsompong K, Intarak N, Kanpittaya P, Trachoo V, Porntaveetus T, Shotelersuk V. Functional consequences of C-terminal mutations in RUNX2. Sci Rep 2023; 13:12202. [PMID: 37500953 PMCID: PMC10374887 DOI: 10.1038/s41598-023-39293-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/22/2023] [Indexed: 07/29/2023] Open
Abstract
Cleidocranial dysplasia (CCD) is a genetic disorder caused by mutations in the RUNX2 gene, affecting bone and teeth development. Previous studies focused on mutations in the RUNX2 RHD domain, with limited investigation of mutations in the C-terminal domain. This study aimed to investigate the functional consequences of C-terminal mutations in RUNX2. Eight mutations were analyzed, and their effects on transactivation activity, protein expression, subcellular localization, and osteogenic potential were studied. Truncating mutations in the PST region and a missense mutation in the NMTS region resulted in increased transactivation activity, while missense mutations in the PST showed activity comparable to the control. Truncating mutations produced truncated proteins, while missense mutations produced normal-sized proteins. Mutant proteins were mislocalized, with six mutant proteins detected in both the nucleus and cytoplasm. CCD patient bone cells exhibited mislocalization of RUNX2, similar to the generated mutant. Mislocalization of RUNX2 and reduced expression of downstream genes were observed in MSCs from a CCD patient with the p.Ser247Valfs*3 mutation, leading to compromised osteogenic potential. This study provides insight into the functional consequences of C-terminal mutations in RUNX2, including reduced expression, mislocalization, and aberrant transactivation of downstream genes, contributing to the compromised osteogenic potential observed in CCD.
Collapse
Affiliation(s)
- Sermporn Thaweesapphithak
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Graduate Program in Oral Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thanakorn Theerapanon
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Khanti Rattanapornsompong
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Narin Intarak
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pimsiri Kanpittaya
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vorapat Trachoo
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thantrira Porntaveetus
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
- Graduate Program in Geriatric and Special Patients Care, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| |
Collapse
|
36
|
Xu X, Li X, Qiu S, Zhou Y, Li L, Chen X, Zheng K, Xu Y. Concentration Selection of Biofriendly Enzyme-Modified Gelatin Hydrogels for Periodontal Bone Regeneration. ACS Biomater Sci Eng 2023; 9:4341-4355. [PMID: 37294274 DOI: 10.1021/acsbiomaterials.3c00166] [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] [Indexed: 06/10/2023]
Abstract
Periodontitis is challenging to cure radically due to its complex periodontal structure and particular microenvironment of dysbiosis and inflammation. However, with the assistance of various materials, cell osteogenic differentiation could be improved, and the ability of hard tissue regeneration could be enhanced. This study aimed to explore the appropriate concentration ratio of biofriendly transglutaminase-modified gelatin hydrogels for promoting periodontal alveolar bone regeneration. Through a series of characterization and cell experiments, we found that all the hydrogels possessed multi-space network structures and demonstrated their biocompatibility. In vivo and in vitro osteogenic differentiation experiments also confirmed that the group 40-5 (transglutaminase-gelatin concentration ratio) possessed a favorable osteogenic potential. In summary, we conclude that such hydrogel with a 40-5 concentration is most conducive to promoting periodontal bone reconstruction, which might be a new route to deal with the dilemma of clinical periodontal treatment.
Collapse
Affiliation(s)
- Xuanwen Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Xinyu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Shuang Qiu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Yi Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Lu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Xu Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Kai Zheng
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Yan Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China
- Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| |
Collapse
|
37
|
Zubaidah N, Pratiwi DD, Masa MMSN, Setiawatie EM, Kunarti S. The Osteogenesis Mechanisms of Dental Alveolar Bone Socket Post Induction with Hydroxyapatite Bovine Tooth Graft: An Animal Experimental in Rattus norvegicus Strain Wistar. Eur J Dent 2023; 17:871-880. [PMID: 36307116 PMCID: PMC10569859 DOI: 10.1055/s-0042-1756691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
OBJECTIVES Surgical endodontics (hemisection) commonly involves the alveolar bone socket and the periradicular tissue. In today's era, optimizing the bone healing process is updated by using bone graft induction. This study explores the mechanisms of bone healing of the alveolar bone socket post-dental extraction of Wistar rats after administration of a bovine tooth graft (hydroxyapatite bovine tooth graft [HAp-BTG]). MATERIALS AND METHODS Fifty Wistar rats were randomly selected into two groups, control and treatment, and into five subgroups on days 3, 7, 14, 21, and 28. The postextraction socket was filled with polyethylene glycol (PEG) as the control and PEG + HAp-BTG as the treatment group. On days 3, 7, 14, 21, and 28, Wistar rats were sacrificed, mandibles were taken, paraffin blocks were made, cut 4 µm thick, and made into glass preparations for microscopic examination. The variable analysis was performed by staining hematoxylin-eosin for osteoblasts (OBs) and osteoclasts (OCs) and immunohistochemistry for runt-related transcription factor 2 (RUNX2), osterix (OSX), osteocalcin (OCN), bone morphogenic protein (BMP) 2. We analyzed the expressed cell count per microscope field. RESULTS In general, the number of cell expressions in the treatment group was significantly higher and faster, except for significantly lower OC. The high variables peak occurred on day 14 for RUNX2 and OCN, on day 7 for OSX, while OB significantly increased on day 21 and remained until day 28. The decrease of OC cells occurred on day 7 and remained low until 28 days. BMP2 was first dominantly induced by HAp-BTG, then the others. CONCLUSION HAp-BTG can induce higher and faster bone healing biomarkers. BMP2 is the dominant first impacted. On the 28th day, it did not significantly express the suppression of OC by OB, which entered the bone formation and remodeling step.
Collapse
Affiliation(s)
- Nanik Zubaidah
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Indonesia
- Department of Conservative Dentistry, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Dian Dwi Pratiwi
- Post Graduate Program of Conservative Dentistry Specialist, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Maria Margaretha S. Nogo Masa
- Post Graduate Program of Conservative Dentistry Specialist, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | | - Sri Kunarti
- Department of Conservative Dentistry, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
38
|
Vimalraj S, Sekaran S. RUNX Family as a Promising Biomarker and a Therapeutic Target in Bone Cancers: A Review on Its Molecular Mechanism(s) behind Tumorigenesis. Cancers (Basel) 2023; 15:3247. [PMID: 37370857 DOI: 10.3390/cancers15123247] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The transcription factor runt-related protein (RUNX) family is the major transcription factor responsible for the formation of osteoblasts from bone marrow mesenchymal stem cells, which are involved in bone formation. Accumulating evidence implicates the RUNX family for its role in tumor biology and cancer progression. The RUNX family has been linked to osteosarcoma via its regulation of many tumorigenicity-related factors. In the regulatory network of cancers, with numerous upstream signaling pathways and its potential target molecules downstream, RUNX is a vital molecule. Hence, a pressing need exists to understand the precise process underpinning the occurrence and prognosis of several malignant tumors. Until recently, RUNX has been regarded as one of the therapeutic targets for bone cancer. Therefore, in this review, we have provided insights into various molecular mechanisms behind the tumorigenic role of RUNX in various important cancers. RUNX is anticipated to grow into a novel therapeutic target with the in-depth study of RUNX family-related regulatory processes, aid in the creation of new medications, and enhance clinical efficacy.
Collapse
Affiliation(s)
- Selvaraj Vimalraj
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Saravanan Sekaran
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, Tamil Nadu, India
| |
Collapse
|
39
|
Lu CW, Lee CJ, Hsieh YJ, Hsu BG. Empagliflozin Attenuates Vascular Calcification in Mice with Chronic Kidney Disease by Regulating the NFR2/HO-1 Anti-Inflammatory Pathway through AMPK Activation. Int J Mol Sci 2023; 24:10016. [PMID: 37373164 DOI: 10.3390/ijms241210016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Vascular calcification (VC) is associated with increased cardiovascular risks in patients with chronic kidney disease (CKD). Sodium-glucose cotransporter 2 inhibitors, such as empagliflozin, can improve cardiovascular and renal outcomes. We assessed the expression of Runt-related transcription factor 2 (Runx2), interleukin (IL)-1β, IL-6, AMP-activated protein kinase (AMPK), nuclear factor erythroid-2-related factor (Nrf2), and heme oxygenase 1 (HO-1) in inorganic phosphate-induced VC in mouse vascular smooth muscle cells (VSMCs) to investigate the mechanisms underlying empagliflozin's therapeutic effects. We evaluated biochemical parameters, mean artery pressure (MAP), pulse wave velocity (PWV), transcutaneous glomerular filtration rate (GFR), and histology in an in vivo mouse model with VC induced by an oral high-phosphorus diet following a 5/6 nephrectomy in ApoE-/- mice. Compared to the control group, empagliflozin-treated mice showed significant reductions in blood glucose, MAP, PWV, and calcification, as well as increased calcium and GFR levels. Empagliflozin inhibited osteogenic trans-differentiation by decreasing inflammatory cytokine expression and increasing AMPK, Nrf2, and HO-1 levels. Empagliflozin mitigates high phosphate-induced calcification in mouse VSMCs through the Nrf2/HO-1 anti-inflammatory pathway by activating AMPK. Animal experiments suggested that empagliflozin reduces VC in CKD ApoE-/- mice on a high-phosphate diet.
Collapse
Affiliation(s)
- Chia-Wen Lu
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan
| | - Chung-Jen Lee
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien 97005, Taiwan
| | - Yi-Jen Hsieh
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
| | - Bang-Gee Hsu
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan
| |
Collapse
|
40
|
Jo YK, Choi B, Zhou C, Jun SH, Cha HJ. Cell recognitive bioadhesive-based osteogenic barrier coating with localized delivery of bone morphogenetic protein-2 for accelerated guided bone regeneration. Bioeng Transl Med 2023; 8:e10493. [PMID: 37206209 PMCID: PMC10189428 DOI: 10.1002/btm2.10493] [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: 08/07/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Titanium mesh (Ti-mesh) for guided bone regeneration (GBR) approaches has been extensively considered to offer space maintenance in reconstructing the alveolar ridge within bone defects due to its superb mechanical properties and biocompatibility. However, soft tissue invasion across the pores of the Ti-mesh and intrinsically limited bioactivity of the titanium substrates often hinder satisfactory clinical outcomes in GBR treatments. Here, a cell recognitive osteogenic barrier coating was proposed using a bioengineered mussel adhesive protein (MAP) fused with Alg-Gly-Asp (RGD) peptide to achieve highly accelerated bone regeneration. The fusion bioadhesive MAP-RGD exhibited outstanding performance as a bioactive physical barrier that enabled effective cell occlusion and a prolonged, localized delivery of bone morphogenetic protein-2 (BMP-2). The MAP-RGD@BMP-2 coating promoted in vitro cellular behaviors and osteogenic commitments of mesenchymal stem cells (MSCs) via the synergistic crosstalk effects of the RGD peptide and BMP-2 in a surface-bound manner. The facile gluing of MAP-RGD@BMP-2 onto the Ti-mesh led to a distinguishable acceleration of the in vivo formation of new bone in terms of quantity and maturity in a rat calvarial defect. Hence, our protein-based cell recognitive osteogenic barrier coating can be an excellent therapeutic platform to improve the clinical predictability of GBR treatment.
Collapse
Affiliation(s)
- Yun Kee Jo
- Department of Biomedical Convergence Science and TechnologySchool of Convergence, Kyungpook National UniversityDaeguRepublic of Korea
- Cell and Matrix Research Institute, Kyungpook National UniversityDaeguSouth Korea
| | | | - Cong Zhou
- School of Stomatology, Shandong UniversityJinanChina
| | - Sang Ho Jun
- Department of Oral and Maxillofacial SurgeryKorea University Anam HospitalSeoulRepublic of Korea
| | - Hyung Joon Cha
- Department of Chemical EngineeringPohang University of Science and TechnologyPohangRepublic of Korea
| |
Collapse
|
41
|
Si W, Kan C, Zhang L, Li F. Role of RUNX2 in breast cancer development and drug resistance (Review). Oncol Lett 2023; 25:176. [PMID: 37033103 PMCID: PMC10079821 DOI: 10.3892/ol.2023.13762] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/02/2023] [Indexed: 03/17/2023] Open
Abstract
Breast cancer is the most common malignancy and ranks second among the causes of tumor-associated death in females. The recurrence and drug resistance of breast cancer are intractable due to the presence of breast cancer stem cells (BCSCs), which are adequate to initiate tumor formation and refractory to conventional remedies. Runt-related transcription factor 2 (RUNX2), a pivotal transcription factor in mammary gland and bone development, has also been related to metastatic cancer and BCSCs. State-of-the-art research has indicated the retention of RUNX2 expression in a more invasive subtype of breast cancer, and in particular, triple-negative breast cancer development and drug resistance are associated with estrogen receptor signaling pathways. The present review mainly focused on the latest updates on RUNX2 in BCSCs and their roles in breast cancer progression and drug resistance, providing insight that may aid the development of RUNX2-based diagnostics and treatments for breast cancer in clinical practice.
Collapse
Affiliation(s)
- Wentao Si
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Chen Kan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Leisheng Zhang
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province and NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
- Key Laboratory of Radiation Technology and Biophysics, Hefei Institute of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China
| | - Feifei Li
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
42
|
Loh HY, Norman BP, Lai KS, Cheng WH, Nik Abd Rahman NMA, Mohamed Alitheen NB, Osman MA. Post-Transcriptional Regulatory Crosstalk between MicroRNAs and Canonical TGF-β/BMP Signalling Cascades on Osteoblast Lineage: A Comprehensive Review. Int J Mol Sci 2023; 24:ijms24076423. [PMID: 37047394 PMCID: PMC10094338 DOI: 10.3390/ijms24076423] [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: 11/07/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 04/14/2023] Open
Abstract
MicroRNAs (miRNAs) are a family of small, single-stranded, and non-protein coding RNAs about 19 to 22 nucleotides in length, that have been reported to have important roles in the control of bone development. MiRNAs have a strong influence on osteoblast differentiation through stages of lineage commitment and maturation, as well as via controlling the activities of osteogenic signal transduction pathways. Generally, miRNAs may modulate cell stemness, proliferation, differentiation, and apoptosis by binding the 3'-untranslated regions (3'-UTRs) of the target genes, which then can subsequently undergo messenger RNA (mRNA) degradation or protein translational repression. MiRNAs manage the gene expression in osteogenic differentiation by regulating multiple signalling cascades and essential transcription factors, including the transforming growth factor-beta (TGF-β)/bone morphogenic protein (BMP), Wingless/Int-1(Wnt)/β-catenin, Notch, and Hedgehog signalling pathways; the Runt-related transcription factor 2 (RUNX2); and osterix (Osx). This shows that miRNAs are essential in regulating diverse osteoblast cell functions. TGF-βs and BMPs transduce signals and exert diverse functions in osteoblastogenesis, skeletal development and bone formation, bone homeostasis, and diseases. Herein, we highlighted the current state of in vitro and in vivo research describing miRNA regulation on the canonical TGF-β/BMP signalling, their effects on osteoblast linage, and understand their mechanism of action for the development of possible therapeutics. In this review, particular attention and comprehensive database searches are focused on related works published between the years 2000 to 2022, using the resources from PubMed, Google Scholar, Scopus, and Web of Science.
Collapse
Affiliation(s)
- Hui-Yi Loh
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Brendan P Norman
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Kok-Song Lai
- Health Sciences Division, Abu Dhabi Women's College, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Wan-Hee Cheng
- Faculty of Health and Life Sciences, INTI International University, Persiaran Perdana BBN, Putra Nilai, Nilai 71800, Negeri Sembilan, Malaysia
| | - Nik Mohd Afizan Nik Abd Rahman
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Noorjahan Banu Mohamed Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Mohd Azuraidi Osman
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
43
|
Accumulation of NCOA1 dependent on HERC3 deficiency transactivates matrix metallopeptidases and promotes extracellular matrix degradation in intervertebral disc degeneration. Life Sci 2023; 320:121555. [PMID: 36878279 DOI: 10.1016/j.lfs.2023.121555] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Matrix metallopeptidases (MMPs) are critical matrix-degrading molecules and they are frequently overexpressed in degenerative discs. This study aimed to investigate the mechanism for MMP upregulation. METHODS Immunoblot and RT-qPCR were used for detecting protein and gene expression levels. 4-month-old and 24-month-old C57BL/6 mice were used for evaluating intervertebral disc degeneration (IDD). An ubiquitination assay was used to determine protein modification. Immunoprecipitation and mass spectrometry were used for identifying protein complex members. RESULTS We identified the elevation of 14 MMPs among 23 members in aged mice with IDD. Eleven of these 14 MMP gene promoters contained a Runx2 (runt-related transcription factor 2) binding site. Biochemical analyses revealed that Runx2 recruited a histone acetyltransferase p300 and a coactivator NCOA1 (nuclear receptor coactivator 1) to assemble a complex, transactivating MMP expression. The deficiency of an E3 ligase called HERC3 (HECT and RLD domain containing E3 ubiquitin-protein ligase 3) resulted in the accumulation of NCOA1 in the inflammatory microenvironment. High throughput screening of small molecules that specifically target the NCOA1-p300 interaction identified a compound SMTNP-191, which showed an inhibitory effect on suppressing MMP expression and attenuating the IDD process in aged mice. CONCLUSION Our data support a model in which deficiency of HERC3 fails to ubiquitinate NCOA1, leading to the assembly of NCOA1-p300-Runx2 and causing the transactivation of MMPs. These findings offer new insight into inflammation-mediated MMP accumulation and also provide a new therapeutic strategy to retard the IDD process.
Collapse
|
44
|
Lorusso F, Scarano A, Fulle S, Valbonetti L, Mancinelli R, Di Filippo ES. Effectiveness of Apigenin, Resveratrol, and Curcumin as Adjuvant Nutraceuticals for Calvarial Bone Defect Healing: An In Vitro and Histological Study on Rats. Nutrients 2023; 15:nu15051235. [PMID: 36904236 PMCID: PMC10005597 DOI: 10.3390/nu15051235] [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: 01/25/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Bone healing is a major clinical issue, especially in bone defects of critical dimensions. Some studies have reported in vivo positive effects on bone healing by some bioactive compounds, such as the phenolic derivatives found in vegetables and plants, such as resveratrol, curcumin, and apigenin. The aim of this work was (1) to analyze in vitro in human dental pulp stem cells the effects of these three natural compounds on the gene expression of related genes downstream to RUNX2 and SMAD5, key factor transcriptions associated with osteoblast differentiation, in order to better understand the positive effects that can occur in vivo in bone healing, and (2) to evaluate in vivo the effects on bone healing of critical-size defects in the calvaria in rats of these three nutraceuticals tested in parallel and for the first time administered by the gastric route. Upregulation of the RUNX2, SMAD5, COLL1, COLL4, and COLL5 genes in the presence of apigenin, curcumin, and resveratrol was detected. In vivo, apigenin induced more consistent significant bone healing in critical-size defects in rat calvaria compared to the other study groups. The study findings encourage a possible therapeutic supplementation with nutraceuticals during the bone regeneration process.
Collapse
Affiliation(s)
- Felice Lorusso
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
- Correspondence: (F.L.); (R.M.)
| | - Antonio Scarano
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Stefania Fulle
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Luca Valbonetti
- Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy
| | - Rosa Mancinelli
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Correspondence: (F.L.); (R.M.)
| | - Ester Sara Di Filippo
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
45
|
Regulon active landscape reveals cell development and functional state changes of human primary osteoblasts in vivo. Hum Genomics 2023; 17:11. [PMID: 36793138 PMCID: PMC9930257 DOI: 10.1186/s40246-022-00448-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 12/20/2022] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND While transcription factor (TF) regulation is known to play an important role in osteoblast development, differentiation, and bone metabolism, the molecular features of TFs in human osteoblasts at the single-cell resolution level have not yet been characterized. Here, we identified modules (regulons) of co-regulated genes by applying single-cell regulatory network inference and clustering to the single-cell RNA sequencing profiles of human osteoblasts. We also performed cell-specific network (CSN) analysis, reconstructed regulon activity-based osteoblast development trajectories, and validated the functions of important regulons both in vivo and in vitro. RESULTS We identified four cell clusters: preosteoblast-S1, preosteoblast-S2, intermediate osteoblasts, and mature osteoblasts. CSN analysis results and regulon activity-based osteoblast development trajectories revealed cell development and functional state changes of osteoblasts. CREM and FOSL2 regulons were mainly active in preosteoblast-S1, FOXC2 regulons were mainly active in intermediate osteoblast, and RUNX2 and CREB3L1 regulons were most active in mature osteoblasts. CONCLUSIONS This is the first study to describe the unique features of human osteoblasts in vivo based on cellular regulon active landscapes. Functional state changes of CREM, FOSL2, FOXC2, RUNX2, and CREB3L1 regulons regarding immunity, cell proliferation, and differentiation identified the important cell stages or subtypes that may be predominantly affected by bone metabolism disorders. These findings may lead to a deeper understanding of the mechanisms underlying bone metabolism and associated diseases.
Collapse
|
46
|
Ghahri T, Salehi Z, Aghajanpour S, Eslaminejad MB, Kalantari N, Akrami M, Dinarvand R, Jang HL, Esfandyari-Manesh M. Development of osteon-like scaffold-cell construct by quadruple coaxial extrusion-based 3D bioprinting of nanocomposite hydrogel. BIOMATERIALS ADVANCES 2023; 145:213254. [PMID: 36584583 DOI: 10.1016/j.bioadv.2022.213254] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/05/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Despite advances in bone tissue engineering, fabricating a scaffold which can be used as an implant for large bone defects remains challenge. One of the great importance in fabricating a biomimetic bone implant is considering the possibility of the integration of the structure and function of implants with hierarchical structure of bone. Herein, we propose a method to mimic the structural unit of compact bone, osteon, with spatial pattern of human umbilical vein endothelial cells (HUVECs) and human mesenchymal stem cells (hMSCs) in the adjacent layers that mimic Haversian canal and lamella, respectively. To this end, coaxial extrusion-based bioprinting technique via a customized quadruple-layer core-shell nozzle was employed. 3D implant scaffold-cell construct was fabricated by using polyethylene glycol as a hollowing agent in the first layer, gelatin methacryloyl (GelMA) and alginate blended hydrogel encapsulating HUVEC cells with vascular endothelial growth factor nanoparticles in the second layer (vasculogenic layer) to mimic vascular vessel, and GelMA and alginate blended hydrogel containing hMSCs cells in the outer osteogenic layer to imitate lamella. Two types of bone minerals, whitlockite and hydroxyapatite, were incorporated in osteogenic layer to induce osteoblastic differentiation and enhance mechanical properties (the young's modules of nanocomposite increased from 35 kPa to 80 kPa). In-vitro evaluations demonstrated high cell viability (94 % within 10 days) and proliferation. Furthermore, ALP enzyme activity increased considerably within 2 weeks and mineralized extra cellular matrix considerably produced within 3 weeks. Also, a significant increase in osteogenic markers was observed indicating the presence of differentiated osteoblast cells. Therefore, the work indicates the potential of single step 3D bioprinting process to fabricate biomimetic osteons to use as bone grafts for regeneration.
Collapse
Affiliation(s)
- Tahmineh Ghahri
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Biotechnology Engineering, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Zeinab Salehi
- Department of Biotechnology Engineering, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Sareh Aghajanpour
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Kalantari
- Department of Stem Cells and Developmental Biology, Royan institute, Tehran, Iran
| | - Mohammad Akrami
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Institute of Biomaterials, University of Tehran & Tehran University of Medical Sciences (IBUTUMS), Tehran, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Leicester School of Pharmacy, De Montfort University, Leicester, UK.
| | - Hae Lin Jang
- Center for Engineered Therapeutics, Department of Medicine and Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Mehdi Esfandyari-Manesh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Medicine, Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
47
|
Tseng C, Han Y, Lv Z, Song Q, Wang K, Shen H, Chen Z. Glucose-stimulated PGC-1α couples with CBP and Runx2 to mediate intervertebral disc degeneration through transactivation of ADAMTS4/5 in diet-induced obesity mice. Bone 2023; 167:116617. [PMID: 36403758 DOI: 10.1016/j.bone.2022.116617] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
Emerging evidence suggests that type 2 diabetes mellitus (T2DM) is associated with the pathogenesis of intervertebral disc degeneration (IDD). However, it is still unclear how T2DM contributes to IDD. Herein, we observed the accumulation of blood glucose and degenerative lumbar discs in mice fed a high-fat diet. Detection of differentially expressed genes in degenerative lumbar discs revealed that ADAMTS4 (A Disintegrin and Metalloproteinase with Thrombospondin motifs) and ADAMTS5 genes were significantly increased. In vitro analyses demonstrated that Runt-Related Transcription Factor 2 (Runx2) recruited both PPARgamma Coactivator 1alpha (PGC-1α) and CREB-Binding Protein (CBP) to transactivate the expression of ADAMTS4/5. Glucose stimulation could dose-dependently induce the accumulation of PGC-1α and promoted the binding of the CBP-PGC-1α-Runx2 complex to the promoters of ADAMTS4/5. Depletion of CBP-PGC-1α-Runx2 complex members and treatment with either PGC-1α inhibitor SR-18292 or CBP inhibitor EML425 in vitro could dramatically inhibit the glucose-induced expression of ADAMTS4/5. Administration of SR-18292 and EML425 in diabetic mice could prevent the degeneration of lumbar discs. Collectively, our results revealed a molecular mechanism by which the hyperglycemia-dependent CBP-PGC-1α-Runx2 complex was required for the transactivation of ADAMTS4/5. The blockage of this complex in diabetic mice may help prevent IDD.
Collapse
Affiliation(s)
- Changchun Tseng
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yingchao Han
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhendong Lv
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qingxin Song
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Wang
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongxing Shen
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Zhi Chen
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
48
|
Ciosek Ż, Kot K, Rotter I. Iron, Zinc, Copper, Cadmium, Mercury, and Bone Tissue. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2197. [PMID: 36767564 PMCID: PMC9915283 DOI: 10.3390/ijerph20032197] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
The paper presents the current understanding on the effects of five metals on bone tissue, namely iron, zinc, copper, cadmium, and mercury. Iron, zinc, and copper contribute significantly to human and animal metabolism when present in sufficient amounts, but their excess or shortage increases the risk of developing bone disorders. In contrast, cadmium and mercury serve no physiological purpose and their long-term accumulation damages the osteoarticular system. We discuss the methods of action and interactions between the discussed elements as well as the concentrations of each element in distinct bone structures.
Collapse
Affiliation(s)
- Żaneta Ciosek
- Chair and Department of Medical Rehabilitation and Clinical Physiotherapy, Pomeranian Medical University in Szczecin, Żołnierska 54, 70-210 Szczecin, Poland
| | - Karolina Kot
- Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Iwona Rotter
- Chair and Department of Medical Rehabilitation and Clinical Physiotherapy, Pomeranian Medical University in Szczecin, Żołnierska 54, 70-210 Szczecin, Poland
| |
Collapse
|
49
|
Bläsius FM, Greven J, Guo W, Bolierakis E, He Z, Lübke C, Simon TP, Hildebrand F, Horst K. Local YB-1, Epo, and EpoR concentrations in fractured bones: results from a porcine model of multiple trauma. Eur J Med Res 2023; 28:25. [PMID: 36639666 PMCID: PMC9837984 DOI: 10.1186/s40001-023-00996-w] [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: 04/25/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
Little is known about the impact of multiple trauma (MT)-related systemic hypoxia on osseous protein concentration of the hypoxia transcriptome. To shed light on this issue, we investigated erythropoietin (Epo), erythropoietin receptor (EpoR), and Y-box binding protein 1 (YB-1) concentrations in the fracture zone in a porcine MT + traumatic hemorrhage (TH) model. Sixteen male domestic pigs were randomized into two groups: an MT + TH group and a sham group. A tibia fracture, lung contusion, and TH were induced in the MT + TH group. The total observation period was 72 h. YB-1 concentrations in bone marrow (BM) were significantly lower in the fracture zone of the MT + TH animals than in the sham animals. Significant downregulation of BM-localized EpoR concentration in both unfractured and fractured bones was observed in the MT + TH animals relative to the sham animals. In BM, Epo concentrations were higher in the fracture zone of the MT + TH animals compared with that in the sham animals. Significantly higher Epo concentrations were detected in the BM of fractured bone compared to that in cortical bone. Our results provide the first evidence that MT + TH alters hypoxia-related protein concentrations. The impacts of both the fracture and concomitant injuries on protein concentrations need to be studied in more detail to shed light on the hypoxia transcriptome in fractured and healthy bones after MT + TH.
Collapse
Affiliation(s)
- Felix Marius Bläsius
- grid.1957.a0000 0001 0728 696XDeptartment of Orthopaedics, Trauma and Reconstructive Surgery, University Hospital, RWTH University, Pauwelsstraße 30, 52074 Aachen, Germany ,grid.1957.a0000 0001 0728 696XInsitute of Pharmacology and Toxicology, University Hospital, RWTH University, Aachen, Germany
| | - Johannes Greven
- grid.1957.a0000 0001 0728 696XDeptartment of Orthopaedics, Trauma and Reconstructive Surgery, University Hospital, RWTH University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Weijun Guo
- grid.1957.a0000 0001 0728 696XDeptartment of Orthopaedics, Trauma and Reconstructive Surgery, University Hospital, RWTH University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Eftychios Bolierakis
- grid.1957.a0000 0001 0728 696XDeptartment of Orthopaedics, Trauma and Reconstructive Surgery, University Hospital, RWTH University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Zhizhen He
- grid.1957.a0000 0001 0728 696XDeptartment of Orthopaedics, Trauma and Reconstructive Surgery, University Hospital, RWTH University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Cavan Lübke
- grid.1957.a0000 0001 0728 696XDepartment of Intensive Care and Intermediate Care, University Hospital, RWTH University, Aachen, Germany
| | - Tim-Philipp Simon
- grid.1957.a0000 0001 0728 696XDepartment of Intensive Care and Intermediate Care, University Hospital, RWTH University, Aachen, Germany
| | - Frank Hildebrand
- grid.1957.a0000 0001 0728 696XDeptartment of Orthopaedics, Trauma and Reconstructive Surgery, University Hospital, RWTH University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Klemens Horst
- grid.1957.a0000 0001 0728 696XDeptartment of Orthopaedics, Trauma and Reconstructive Surgery, University Hospital, RWTH University, Pauwelsstraße 30, 52074 Aachen, Germany
| |
Collapse
|
50
|
Jiang L, Lu Y, Zhao H, He W. Polysaccharides from aloe vera target the Wnt/β-catenin pathway to impact the tooth density of pulpitis rats. Acta Cir Bras 2023; 37:e371202. [PMID: 36651427 PMCID: PMC9839156 DOI: 10.1590/acb371202] [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: 08/09/2022] [Accepted: 11/06/2022] [Indexed: 01/14/2023] Open
Abstract
PURPOSE To investigate the mechanism of polysaccharides from aloe vera (PAV), a main active ingredient of Aloe vera, treatment in pulpitis rats. METHODS Pulpitis were modeled by drilling the occlusal central fossa with Sprague Dawley rats. Next, the rats were treated with 20, 40, and 80 mg/kg PAV for three weeks, respectively. Computed tomography scanning assay, hematoxylin and eosin staining, and tartrate-resistant acid phosphatase staining were used to detect the pathology change. Then, levels of tumor necrosis factor-α, interleukin-1β, prostaglandin E2, and ciclooxigenase 2 were detected by enzyme-linked immunosorbent assay. The expressions of bone morphogenetic protein 2 human (BMP-2), osteocalcin, osterix, and runt-related transcription factor 2 (Runx2) were quantified by quantitative real-time polymerase chain reaction and Western blotting (WB). Finally, Wnt3a expression, p-GSK3β/GSK3β and p-β-catenin/β-catenin ratio were analyzed by WB. RESULTS PAV up regulated the bone mineral density, and reduced the breakage of the crown and cervical structures, and the necrosis of the crown and root pulp of pulpitis rats. In addition, results indicated that PAV could inhibit osteoblast formation. While osteoblasts' number was decreased, proteins of BMP-2, osteocalcin, osterix, and Runx2 were up-regulated by PAV. Furthermore, PAV increased the Wnt3a expression and the p-β-catenin/β-catenin ratio, and decreased p-GSK3β/GSK3β ratio. Interestingly, these effects were all in dose dependence. CONCLUSIONS PAV could inhibit pulp inflammation and promote osteoblasts differentiation via suppressing the activation of the Wnt/β-catenin signaling, enhancing the dental bone density.
Collapse
Affiliation(s)
- Ling Jiang
- MD. Chongqing Medical University – Department of Urology Surgery – The First Affiliated Hospital – Chongqing, China
| | - Yang Lu
- MD. Chongqing Medical University – Department of Urology Surgery – The First Affiliated Hospital – Chongqing, China
| | - Hongyan Zhao
- MD. Chongqing University – Department of Plastic Surgery – Central Hospital – Chongqing, China
| | - Weiyang He
- MD. Chongqing Medical University – Department of Urology Surgery – The First Affiliated Hospital – Chongqing, China.,Corresponding author:
- 13051377276
| |
Collapse
|