1
|
Xu G, Zhang W, Yang J, Sun N, Qu X. Identification of neutrophil extracellular traps and crosstalk genes linking inflammatory bowel disease and osteoporosis by integrated bioinformatics analysis and machine learning. Sci Rep 2023; 13:23054. [PMID: 38155235 PMCID: PMC10754907 DOI: 10.1038/s41598-023-50488-4] [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: 08/24/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023] Open
Abstract
Musculoskeletal deficits are among the most common extra-intestinal manifestations and complications of inflammatory bowel disease (IBD). This study aimed to identify crosstalk genes between IBD and osteoporosis (OP) and potential relationships between crosstalk and neutrophil extracellular traps (NETs)-related genes. Three common hub genes from different compared groups are actually the same, namely HDAC6, IL-8, and PPIF. ROC showed that the combined diagnostic value of HDAC6, IL-8, and PPIF was higher than each of the three key hub genes. Immune infiltration results showed that HDAC6 and IL-8 key genes negatively correlated with CD65 bright natural killer cells. USF1 was the common upstream TFs between HDAC6 and PPIF, and MYC was the common upstream TFs between IL-8 and PPIF in RegNetwork. Taken together, this study shows a linked mechanism between IBD and OP via NETs and crosstalk genes. These findings may show light on better diagnosis and treatment of IBD complicated with OP.
Collapse
Affiliation(s)
- Gang Xu
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China.
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases, Dalian, Liaoning Province, China.
| | - Wanhao Zhang
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Jun Yang
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Na Sun
- Department of Pharmacy, The Third People's Hospital of Dalian, Dalian, Liaoning Province, China
| | - Xiaochen Qu
- Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China.
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases, Dalian, Liaoning Province, China.
| |
Collapse
|
2
|
Pofi R, Caratti G, Ray DW, Tomlinson JW. Treating the Side Effects of Exogenous Glucocorticoids; Can We Separate the Good From the Bad? Endocr Rev 2023; 44:975-1011. [PMID: 37253115 PMCID: PMC10638606 DOI: 10.1210/endrev/bnad016] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 06/01/2023]
Abstract
It is estimated that 2% to 3% of the population are currently prescribed systemic or topical glucocorticoid treatment. The potent anti-inflammatory action of glucocorticoids to deliver therapeutic benefit is not in doubt. However, the side effects associated with their use, including central weight gain, hypertension, insulin resistance, type 2 diabetes (T2D), and osteoporosis, often collectively termed iatrogenic Cushing's syndrome, are associated with a significant health and economic burden. The precise cellular mechanisms underpinning the differential action of glucocorticoids to drive the desirable and undesirable effects are still not completely understood. Faced with the unmet clinical need to limit glucocorticoid-induced adverse effects alongside ensuring the preservation of anti-inflammatory actions, several strategies have been pursued. The coprescription of existing licensed drugs to treat incident adverse effects can be effective, but data examining the prevention of adverse effects are limited. Novel selective glucocorticoid receptor agonists and selective glucocorticoid receptor modulators have been designed that aim to specifically and selectively activate anti-inflammatory responses based upon their interaction with the glucocorticoid receptor. Several of these compounds are currently in clinical trials to evaluate their efficacy. More recently, strategies exploiting tissue-specific glucocorticoid metabolism through the isoforms of 11β-hydroxysteroid dehydrogenase has shown early potential, although data from clinical trials are limited. The aim of any treatment is to maximize benefit while minimizing risk, and within this review we define the adverse effect profile associated with glucocorticoid use and evaluate current and developing strategies that aim to limit side effects but preserve desirable therapeutic efficacy.
Collapse
Affiliation(s)
- Riccardo Pofi
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Giorgio Caratti
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - David W Ray
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Oxford Kavli Centre for Nanoscience Discovery, University of Oxford, Oxford OX37LE, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| |
Collapse
|
3
|
Nguyen TT, Phuong MT, Shrestha M, Park J, Le Minh P, Kim JO, Choi H, Nam JW, Jiang HL, Kim HS, Jeong JH, Park JB, Yook S. Scalable and Uniform Fabrication of Dexamethasone-Eluting Depot-Engineered Stem Cell Spheroids as a Microtissue Construct to Target Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:26373-26384. [PMID: 37219569 DOI: 10.1021/acsami.3c03102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Potentiation of stem cell potency is critical for successful tissue engineering, especially for bone regeneration. Three-dimensional cell culture and bioactive molecule co-delivery with cells have been proposed to achieve this effect. Here, we provide a uniform and scalable fabrication of osteogenic microtissue constructs of mesenchymal stem cell (MSC) spheroids surface-engineered with dexamethasone-releasing polydopamine-coated microparticles (PD-DEXA/MPs) to target bone regeneration. The microparticle conjugation process was rapid and cell-friendly and did not affect the cell viability or key functionalities. The incorporation of DEXA in the conjugated system significantly enhanced the osteogenic differentiation of MSC spheroids, as evidenced by upregulating osteogenic gene expression and intense alkaline phosphatase and alizarin red S staining. In addition, the migration of MSCs from spheroids was tested on a biocompatible macroporous fibrin scaffold (MFS). The result showed that PD-DEXA/MPs were stably anchored on MSCs during cell migration over time. Finally, the implantation of PD-DEXA/MP-conjugated spheroid-loaded MFS into a calvarial defect in a mouse model showed substantial bone regeneration. In conclusion, the uniform fabrication of microtissue constructs containing MSC spheroids with drug depots shows a potential to improve the performance of MSCs in tissue engineering.
Collapse
Affiliation(s)
- Tiep Tien Nguyen
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| | - Mai Thi Phuong
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| | - Manju Shrestha
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Junhyeung Park
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Pham Le Minh
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| | - Hyukjae Choi
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| | - Joo-Won Nam
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China
| | - Hyung-Sik Kim
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
- Dental and Life Science Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
4
|
Wang Z, Wen S, Zhong M, Yang Z, Xiong W, Zhang K, Yang S, Li H, Guo S. Epigenetics: Novel crucial approach for osteogenesis of mesenchymal stem cells. J Tissue Eng 2023; 14:20417314231175364. [PMID: 37342486 PMCID: PMC10278427 DOI: 10.1177/20417314231175364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/26/2023] [Indexed: 06/23/2023] Open
Abstract
Bone has a robust regenerative potential, but its capacity to repair critical-sized bone defects is limited. In recent years, stem cells have attracted significant interest for their potential in tissue engineering. Applying mesenchymal stem cells (MSCs) for enhancing bone regeneration is a promising therapeutic strategy. However, maintaining optimal cell efficacy or viability of MSCs is limited by several factors. Epigenetic modification can cause changes in gene expression levels without changing its sequence, mainly including nucleic acids methylation, histone modification, and non-coding RNAs. This modification is believed to be one of the determinants of MSCs fate and differentiation. Understanding the epigenetic modification of MSCs can improve the activity and function of stem cells. This review summarizes recent advances in the epigenetic mechanisms of MSCs differentiation into osteoblast lineages. We expound that epigenetic modification of MSCs can be harnessed to treat bone defects and promote bone regeneration, providing potential therapeutic targets for bone-related diseases.
Collapse
Affiliation(s)
- Zhaohua Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Si Wen
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Meiqi Zhong
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ziming Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wei Xiong
- Department of Plastic Surgery, The First Hospital of Shihezi University School of Medicine, Shihezi, China
| | - Kuo Zhang
- College of Humanities and Social Sciences, Dalian Medical University, Dalian, Liaoning Province, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Huizheng Li
- Department of Otorhinolaryngology & Head and Neck Surgery, Dalian Friendship Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
5
|
Xing F, Yin HM, Zhe M, Xie JC, Duan X, Xu JZ, Xiang Z, Li ZM. Nanotopographical 3D-Printed Poly(ε-caprolactone) Scaffolds Enhance Proliferation and Osteogenic Differentiation of Urine-Derived Stem Cells for Bone Regeneration. Pharmaceutics 2022; 14:pharmaceutics14071437. [PMID: 35890332 PMCID: PMC9317219 DOI: 10.3390/pharmaceutics14071437] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/02/2022] [Accepted: 07/06/2022] [Indexed: 02/05/2023] Open
Abstract
3D-printing technology can be used to construct personalized bone substitutes with customized shapes, but it cannot regulate the topological morphology of the scaffold surface, which plays a vital role in regulating the biological behaviors of stem cells. In addition, stem cells are able to sense the topographical and mechanical cues of surface of scaffolds by mechanosensing and mechanotransduction. In our study, we fabricated a 3D-printed poly(ε-caprolactone) (PCL) scaffold with a nanotopographical surface and loaded it with urine-derived stem cells (USCs) for application of bone regeneration. The topological 3D-printed PCL scaffolds (TPS) fabricated by surface epiphytic crystallization, possessed uniformly patterned nanoridges, of which the element composition and functional groups of nanoridges were the same as PCL. Compared with bare 3D-printed PCL scaffolds (BPS), TPS have a higher ability for protein adsorption and mineralization in vitro. The proliferation, cell length, and osteogenic gene expression of USCs on the surface of TPS were significantly higher than that of BPS. In addition, the TPS loaded with USCs exhibited a good ability for bone regeneration in cranial bone defects. Our study demonstrated that nanotopographical 3D-printed scaffolds loaded with USCs are a safe and effective therapeutic strategy for bone regeneration.
Collapse
Affiliation(s)
- Fei Xing
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China; (F.X.); (Z.X.)
| | - Hua-Mo Yin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China; (H.-M.Y.); (Z.-M.L.)
| | - Man Zhe
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Ji-Chang Xie
- Laboratoire Roberval, FRE UTC-CNRS 2012, Sorbonne Universités, Université de Technologie de Compiègne, Centre de Recherche Royallieu, CS60319, CEDEX, 60203 Compiègne, France;
| | - Xin Duan
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China; (F.X.); (Z.X.)
- Correspondence: (X.D.); (J.-Z.X.)
| | - Jia-Zhuang Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China; (H.-M.Y.); (Z.-M.L.)
- Correspondence: (X.D.); (J.-Z.X.)
| | - Zhou Xiang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China; (F.X.); (Z.X.)
| | - Zhong-Ming Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China; (H.-M.Y.); (Z.-M.L.)
| |
Collapse
|
6
|
Liu S, Zhou Y, Chen Y, Liu Y, Peng S, Cao Z, Xia H. Bmal1 promotes cementoblast differentiation and cementum mineralization via Wnt/β-catenin signaling. Acta Histochem 2022; 124:151868. [PMID: 35183881 DOI: 10.1016/j.acthis.2022.151868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/24/2022] [Accepted: 02/11/2022] [Indexed: 01/09/2023]
Abstract
Remodeling of the cementum plays a crucial role in periodontal regenerative therapy, while the precise mechanism of cementogenesis has yet been adequately understood. Recent studies have indicated the connection between osteogenic differentiation and Brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein-1 (Bmal1). Besides, Wnt/β-catenin signaling is proven to be an essential regulator in cementogenesis. In this study, we found a robust expression of Bmal1 in cementoblasts in the mandibular first molar of mice by immunohistochemical staining. To further explore the role of Bmal1 in cementogenesis, we examined the expression pattern of Bmal1 in OCCM-30, an immortalized murine cementoblast cell line by qRT-PCR and western blot. Our data demonstrated the upregulation of Bmal1 at both mRNA and protein levels during differentiation. Additionally, stable knockdown of Bmal1 in OCCM-30 cells resulted in downregulation of osteogenic markers such as alkaline phosphatase (Alp), osteopontin (Opn), and osteocalcin (Ocn), and reduced formation of mineralized nodules. Moreover, qRT-PCR and western blot results exhibited that the expression of β-catenin was attenuated by Bmal1 deficiency. We also found that the mRNA levels of Tcf1 and Lef1, the target transcription factors of β-catenin, were reduced by Bmal1 deficiency. In conclusion, this study preliminarily confirms that Bmal1 promotes cementoblast differentiation and cementum mineralization via Wnt/β-catenin signaling, which contributes to a potential strategy in periodontal regenerative therapy.
Collapse
Affiliation(s)
- Shumin Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei - MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, People's Republic of China
| | - Yi Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei - MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, People's Republic of China
| | - Yang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei - MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, People's Republic of China
| | - Ying Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei - MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, People's Republic of China
| | - Shuzhen Peng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei - MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, People's Republic of China
| | - Zhengguo Cao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei - MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, People's Republic of China
| | - Haibin Xia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei - MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, People's Republic of China.
| |
Collapse
|
7
|
Hohenbild F, Arango Ospina M, Schmitz SI, Moghaddam A, Boccaccini AR, Westhauser F. An In Vitro Evaluation of the Biological and Osteogenic Properties of Magnesium-Doped Bioactive Glasses for Application in Bone Tissue Engineering. Int J Mol Sci 2021; 22:12703. [PMID: 34884519 PMCID: PMC8657676 DOI: 10.3390/ijms222312703] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/14/2021] [Accepted: 11/19/2021] [Indexed: 01/16/2023] Open
Abstract
Magnesium (Mg2+) is known to play a crucial role in mineral and matrix metabolism of bone tissue and is thus increasingly considered in the field of bone tissue engineering. Bioactive glasses (BGs) offer the promising possibility of the incorporation and local delivery of therapeutically active ions as Mg2+. In this study, two Mg2+-doped derivatives of the ICIE16-BG composition (49.46 SiO2, 36.27 CaO, 6.6 Na2O, 1.07 P2O5, 6.6 K2O (mol%)), namely 6Mg-BG (49.46 SiO2, 30.27 CaO, 6.6 Na2O, 1.07 P2O5, 6.6 K2O, 6.0 MgO (mol%) and 3Mg-BG (49.46 SiO2, 33.27 CaO, 6.6 Na2O, 1.07 P2O5, 6.6 K2O, 3.0 MgO (mol%)) were examined. Their influence on viability, proliferation and osteogenic differentiation of human mesenchymal stromal cells (MSCs) was explored in comparison to the original ICIE16-BG. All BGs showed good biocompatibility. The Mg2+-doped BGs had a positive influence on MSC viability alongside with inhibiting effects on MSC proliferation. A strong induction of osteogenic differentiation markers was observed, with the Mg2+-doped BGs significantly outperforming the ICIE16-BG regarding the expression of genes encoding for protein members of the osseous extracellular matrix (ECM) at certain observation time points. However, an overall Mg2+-induced enhancement of the expression of genes encoding for ECM proteins could not be observed, possibly due to a too moderate Mg2+ release. By adaption of the Mg2+ release from BGs, an even stronger impact on the expression of genes encoding for ECM proteins might be achieved. Furthermore, other BG-types such as mesoporous BGs might provide a higher local presence of the therapeutically active ions and should therefore be considered for upcoming studies.
Collapse
Affiliation(s)
- Frederike Hohenbild
- Center of Orthopedics, Traumatology and Spinal Cord Injury, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Germany; (F.H.); (S.I.S.)
| | - Marcela Arango Ospina
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstr. 6, 91058 Erlangen, Germany; (M.A.O.); (A.R.B.)
| | - Sarah I. Schmitz
- Center of Orthopedics, Traumatology and Spinal Cord Injury, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Germany; (F.H.); (S.I.S.)
| | - Arash Moghaddam
- Orthopedic and Trauma Surgery, Frohsinnstraße 12, 63739 Aschaffenburg, Germany;
| | - Aldo R. Boccaccini
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstr. 6, 91058 Erlangen, Germany; (M.A.O.); (A.R.B.)
| | - Fabian Westhauser
- Center of Orthopedics, Traumatology and Spinal Cord Injury, Heidelberg University Hospital, Schlierbacher Landstraße 200a, 69118 Heidelberg, Germany; (F.H.); (S.I.S.)
| |
Collapse
|
8
|
Enhanced osteoinductive capacity of poly(lactic-co-glycolic) acid and biphasic ceramic scaffolds by embedding simvastatin. Clin Oral Investig 2021; 26:2693-2701. [PMID: 34694495 DOI: 10.1007/s00784-021-04240-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/15/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES This study evaluated the effect of embedding simvastatin (SIM) on the osteoinductive capacity of PLGA + HA/βTCP scaffolds in stem cells from human exfoliated deciduous teeth (SHED). MATERIALS AND METHODS Scaffolds were produced by PLGA solvent dissolution, addition of HA/βTCP, solvent evaporation, and leaching of sucrose particles to impart porosity. Biphasic ceramic particles (70% HA/30% βTCP) were added to the PLGA in a 1:1 (w:w) ratio. Scaffolds with SIM received 1% (w:w) of this medication. Scaffolds were synthesized in a disc-shape and sterilized by ethylene oxide. The experimental groups were (G1) PLGA + HA/βTCP and (G2) PLGA + HA/βTCP + SIM in non-osteogenic culture medium, while (G3) SHED and (G4) MC3T3-E1 in osteogenic culture medium were the positive control groups. The release profile of SIM from scaffolds was evaluated. DNA quantification assay, alkaline phosphatase activity, osteocalcin and osteonectin proteins, extracellular calcium detection, von Kossa staining, and X-ray microtomography were performed to assess the capacity of scaffolds to induce the osteogenic differentiation of SHED. RESULTS The release profile of SIM followed a non-liner sustained-release rate, reaching about 40% of drug release at day 28. Additionally, G2 promoted the highest osteogenic differentiation of SHED, even when compared to the positive control groups. CONCLUSIONS In summary, the osteoinductive capacity of poly(lactic-co-glycolic) acid and biphasic ceramic scaffolds was expressively enhanced by embedding simvastatin. CLINICAL RELEVANCE Bone regeneration is still a limiting factor in the success of several approaches to oral and maxillofacial surgeries, though tissue engineering using mesenchymal stem cells, scaffolds, and osteoinductive mediators might collaborate to this topic.
Collapse
|
9
|
Martin-Aragon S, Bermejo-Bescós P, Benedí J, Raposo C, Marques F, Kydonaki EK, Gkiata P, Koutedakis Y, Ntina G, Carrillo AE, Amorim T. A Neuroprotective Bovine Colostrum Attenuates Apoptosis in Dexamethasone-Treated MC3T3-E1 Osteoblastic Cells. Int J Mol Sci 2021; 22:10195. [PMID: 34638536 PMCID: PMC8507997 DOI: 10.3390/ijms221910195] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 01/10/2023] Open
Abstract
Glucocorticoid-induced osteoporosis (GIO) is one of the most common secondary forms of osteoporosis. GIO is partially due to the apoptosis of osteoblasts and osteocytes. In addition, high doses of dexamethasone (DEX), a synthetic glucocorticoid receptor agonist, induces neurodegeneration by initiating inflammatory processes leading to neural apoptosis. Here, a neuroprotective bovine colostrum against glucocorticoid-induced neuronal damage was investigated for its anti-apoptotic activity in glucocorticoid-treated MC3T3-E1 osteoblastic cells. A model of apoptotic osteoblastic cells was developed by exposing MC3T3-E1 cells to DEX (0-700 μM). Colostrum co-treated with DEX was executed at 0.1-5.0 mg/mL. Cell viability was measured for all treatment schedules. Caspase-3 activation was assessed to determine both osteoblast apoptosis under DEX exposure and its potential prevention by colostrum co-treatment. Glutathione reduced (GSH) was measured to determine whether DEX-mediated oxidative stress-driven apoptosis is alleviated by colostrum co-treatment. Western blot was performed to determine the levels of p-ERK1/2, Bcl-XL, Bax, and Hsp70 proteins upon DEX or DEX plus colostrum exposure. Colostrum prevented the decrease in cell viability and the increase in caspase-3 activation and oxidative stress caused by DEX exposure. Cells, upon colostrum co-treated with DEX, exhibited higher levels of p-ERK1/2 and lower levels of Bcl-XL, Bax, and Hsp70. Our data support the notion that colostrum may be able to reduce DEX-induced apoptosis possibly via the activation of the ERK pathway and modulation of the Hsp70 system. We provided preliminary evidence on how bovine colostrum, as a complex and multi-component dairy product, in addition to its neuroprotective action, may affect osteoblastic cell survival undergoing apoptosis.
Collapse
Affiliation(s)
- Sagrario Martin-Aragon
- Department of Pharmacology, Pharmacognosy and Botany, Complutense University, 28040 Madrid, Spain
| | - Paloma Bermejo-Bescós
- Department of Pharmacology, Pharmacognosy and Botany, Complutense University, 28040 Madrid, Spain
| | - Juana Benedí
- Department of Pharmacology, Pharmacognosy and Botany, Complutense University, 28040 Madrid, Spain
| | - Carlos Raposo
- Department of Pharmacology, Pharmacognosy and Botany, Complutense University, 28040 Madrid, Spain
- SALURIS, 28040 Madrid, Spain
| | - Franklim Marques
- UCIBIO/REQUIMTE, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Eirini K Kydonaki
- UCIBIO/REQUIMTE, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Paraskevi Gkiata
- School of Sport and Exercise Sciences, University of Thessaly, Karies, 42100 Trikala, Greece
| | - Yiannis Koutedakis
- School of Sport and Exercise Sciences, University of Thessaly, Karies, 42100 Trikala, Greece
- Faculty of Education, Health and Wellbeing, Wolverhampton University, Walsall WV1 1LY, UK
| | - Georgia Ntina
- BME, Biomechanical Solutions, 43150 Karditsa, Greece
| | - Andres E Carrillo
- Department of Exercise Science, Chatham University, Pittsburgh, PA 15232, USA
- Move-Cor Inc., Pittsburgh, PA 15017, USA
| | - Tânia Amorim
- UCIBIO/REQUIMTE, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| |
Collapse
|
10
|
Della Bella E, Buetti-Dinh A, Licandro G, Ahmad P, Basoli V, Alini M, Stoddart MJ. Dexamethasone Induces Changes in Osteogenic Differentiation of Human Mesenchymal Stromal Cells via SOX9 and PPARG, but Not RUNX2. Int J Mol Sci 2021; 22:4785. [PMID: 33946412 PMCID: PMC8124248 DOI: 10.3390/ijms22094785] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/24/2021] [Accepted: 04/28/2021] [Indexed: 01/10/2023] Open
Abstract
Despite the huge body of research on osteogenic differentiation and bone tissue engineering, the translation potential of in vitro results still does not match the effort employed. One reason might be that the protocols used for in vitro research have inherent pitfalls. The synthetic glucocorticoid dexamethasone is commonly used in protocols for trilineage differentiation of human bone marrow mesenchymal stromal cells (hBMSCs). However, in the case of osteogenic commitment, dexamethasone has the main pitfall of inhibiting terminal osteoblast differentiation, and its pro-adipogenic effect is well known. In this work, we aimed to clarify the role of dexamethasone in the osteogenesis of hBMSCs, with a particular focus on off-target differentiation. The results showed that dexamethasone does induce osteogenic differentiation by inhibiting SOX9 expression, but not directly through RUNX2 upregulation as it is commonly thought. Rather, PPARG is concomitantly and strongly upregulated, leading to the formation of adipocyte-like cells within osteogenic cultures. Limiting the exposure to dexamethasone to the first week of differentiation did not affect the mineralization potential. Gene expression levels of RUNX2, SOX9, and PPARG were simulated using approximate Bayesian computation based on a simplified theoretical model, which was able to reproduce the observed experimental trends but with a different range of responses, indicating that other factors should be integrated to fully understand how dexamethasone influences cell fate. In summary, this work provides evidence that current in vitro differentiation protocols based on dexamethasone do not represent a good model, and further research is warranted in this field.
Collapse
Affiliation(s)
- Elena Della Bella
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; (E.D.B.); (P.A.); (V.B.); (M.A.)
| | - Antoine Buetti-Dinh
- Laboratory of applied microbiology (LMA), Department of Environment, Constructions and Design (DACD), University of Applied Sciences of Southern Switzerland (SUPSI), 6500 Bellinzona, Switzerland;
- Swiss Institute of Bioinformatics, Quartier Sorge—Batiment Genopode, 1015 Lausanne, Switzerland
| | - Ginevra Licandro
- Dalle Molle Institute for Artificial Intelligence (IDSIA), University of Italian Switzerland (USI), 6928 Manno, Switzerland;
- University of Applied Science and Art of Southern Switzerland (SUPSI), 6928 Manno, Switzerland
| | - Paras Ahmad
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; (E.D.B.); (P.A.); (V.B.); (M.A.)
| | - Valentina Basoli
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; (E.D.B.); (P.A.); (V.B.); (M.A.)
| | - Mauro Alini
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; (E.D.B.); (P.A.); (V.B.); (M.A.)
| | - Martin J. Stoddart
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; (E.D.B.); (P.A.); (V.B.); (M.A.)
- Department of Orthopedics and Trauma Surgery, Medical Center—Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
11
|
Zhao H, Yeersheng R, Xia Y, Kang P, Wang W. Hypoxia Enhanced Bone Regeneration Through the HIF-1α/β-Catenin Pathway in Femoral Head Osteonecrosis. Am J Med Sci 2021; 362:78-91. [PMID: 33727018 DOI: 10.1016/j.amjms.2021.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/12/2020] [Accepted: 03/11/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Osteonecrosis of the femoral head (ONFH) is a common disease. Transplantation of bone marrow stem cells (BMSCs) is a promising method to treat ONFH but is impeded by the low survival rate and deficiency of cell bioactivity. METHODS We performed hypoxic preprocessing to treat BMSCs and assessed cell viability, apoptosis, differentiation, and growth factor expression in vitro. Subsequently, we constructed the ONFH model and delivered hypoxia-pretreated BMSCs to the rabbit femoral head after core decompression surgery, evaluating its effects on bone regeneration and ONFH repair. Six weeks later, micro-computed tomography (CT) and histopathology were performed to evaluate ONFH repair. RESULTS Our findings demonstrated that hypoxic preprocessing promoted the viability of BMSCs, increased the expression of hypoxia-inducible factor-1 alpha (HIF-1α), vascular endothelial growth factor (VEGF), alkaline phosphatase (ALP), calcium deposition, and enhanced the formation of vessels-shaped structures. In an in vivo study, micro-CT observations demonstrated that the bone volume was increased in the hypoxia BMSCs group. Histological examination revealed reduced cellular apoptosis, lower empty lacunae rate, enhanced bone formation, and stronger trabecular bone in the hypoxia BMSCs group when compared with those transplanted with normoxia treated BMSCs. Additionally, immunological assessment of the hypoxia BMSCs group demonstrated increased expression of HIF-1α and β-catenin, as well as increased VEGF, ALP, osteocalcin (OCN), and collagen type I (Col-1). CONCLUSIONS Collectively, our findings indicated that hypoxia stimulated angiogenesis and bone regeneration via the HIF-1/β-catenin pathway in BMSCs and that the delivery of hypoxia-pretreated BMSCs contributed to the treatment of early ONFH.
Collapse
Affiliation(s)
- HaiYan Zhao
- Department of Orthopedics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Releken Yeersheng
- Department of Orthopedics, The First Hospital of Lanzhou University, Lanzhou, China
| | - YaYi Xia
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| | - PengDe Kang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - WenJi Wang
- Department of Orthopedics, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
12
|
Gasson SB, Dobson LK, Chow L, Dow S, Gregory CA, Saunders WB. Optimizing In Vitro Osteogenesis in Canine Autologous and Induced Pluripotent Stem Cell-Derived Mesenchymal Stromal Cells with Dexamethasone and BMP-2. Stem Cells Dev 2021; 30:214-226. [PMID: 33356875 PMCID: PMC7891305 DOI: 10.1089/scd.2020.0144] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022] Open
Abstract
A growing body of work suggests that canine mesenchymal stromal cells (cMSCs) require additional agonists such as bone morphogenic protein-2 (BMP-2) for consistent in vitro osteogenic differentiation. BMP-2 is costly and may challenge the translational relevance of the canine model. Dexamethasone enhances osteogenic differentiation of human MSCs (hMSCs) and is widely utilized in osteogenic protocols. The aim of this study was to determine the effect of BMP-2 and dexamethasone on early- and late-stage osteogenesis of autologous and induced pluripotent stem cell (iPS)-derived cMSCs. Two preparations of marrow-derived cMSCs were selected to represent exceptionally or marginally osteogenic autologous cMSCs. iPS-derived cMSCs were generated from canine fibroblasts. All preparations were evaluated using alkaline phosphatase (ALP) activity, Alizarin Red staining of osteogenic monolayers, and quantitative polymerase chain reaction. Data were reported as mean ± standard deviation and compared using one- or two-way analysis of variance and Tukey or Sidak post hoc tests. Significance was established at P < 0.05. In early-stage assays, dexamethasone decreased ALP activity for all cMSCs in the presence of BMP-2. In late-stage assays, inclusion of dexamethasone and BMP-2 at Day 1 of culture produced robust monolayer mineralization for autologous cMSCs. Delivering 100 nM dexamethasone at Day 1 improved mineralization and reduced the BMP-2 concentrations required to achieve mineralization of the marginal cMSCs. For iPS-cMSCs, dexamethasone was inhibitory to both ALP activity and monolayer mineralization. There was increased expression of osteocalcin and osterix with BMP-2 in autologous cMSCs but a more modest expression occurred in iPS cMSCs. While autologous and iPS-derived cMSCs respond similarly in early-stage osteogenic assays, they exhibit unique responses to dexamethasone and BMP-2 in late-stage mineralization assays. This study demonstrates that dexamethasone and BMP-2 can be titrated in a time- and concentration-dependent manner to enhance osteogenesis of autologous cMSC preparations. These results will prove useful for investigators performing translational studies with cMSCs while providing insight into iPS-derived cMSC osteogenesis.
Collapse
Affiliation(s)
- Shelby B. Gasson
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Lauren K. Dobson
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Lyndah Chow
- Department of Clinical Sciences, Center for Immune and Regenerative Medicine, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Steven Dow
- Department of Clinical Sciences, Center for Immune and Regenerative Medicine, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Carl A. Gregory
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - William Brian Saunders
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
13
|
Sordi MB, Curtarelli RB, da Silva IT, Fongaro G, Benfatti CAM, de Souza Magini R, Cabral da Cruz AC. Effect of dexamethasone as osteogenic supplementation in in vitro osteogenic differentiation of stem cells from human exfoliated deciduous teeth. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:1. [PMID: 33469820 PMCID: PMC7815568 DOI: 10.1007/s10856-020-06475-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 12/10/2020] [Indexed: 05/05/2023]
Abstract
In in vitro culture systems, dexamethasone (DEX) has been applied with ascorbic acid (ASC) and β-glycerophosphate (βGLY) as culture media supplementation to induce osteogenic differentiation of mesenchymal stem cells. However, there are some inconsistencies regarding the role of DEX as osteogenic media supplementation. Therefore, this study verified the influence of DEX culture media supplementation on the osteogenic differentiation, especially the capacity to mineralize the extracellular matrix of stem cells from human exfoliated deciduous teeth (SHED). Five groups were established: G1-SHED + Dulbecco's Modified Eagles' Medium (DMEM) + fetal bovine serum (FBS); G2-SHED + DMEM + FBS + DEX; G3-SHED + DMEM + FBS + ASC + βGLY; G4-SHED + DMEM + FBS + ASC + βGLY + DEX; G5-MC3T3-E1 + α Minimal Essential Medium (MEM) + FBS + ASC + βGLY. DNA content, alkaline phosphatase (ALP) activity, free calcium quantification in the extracellular medium, and extracellular matrix mineralization quantification through staining with von Kossa, alizarin red, and tetracycline were performed on days 7 and 21. Osteogenic media supplemented with ASC and β-GLY demonstrated similar effects on SHED in the presence or absence of DEX for DNA content (day 21) and capacity to mineralize the extracellular matrix according to alizarin red and tetracycline quantifications (day 21). In addition, the presence of DEX in the osteogenic medium promoted less ALP activity (day 7) and extracellular matrix mineralization according to the von Kossa assay (day 21), and more free calcium quantification at extracellular medium (day 21). In summary, the presence of DEX in the osteogenic media supplementation did not interfere with SHED commitment into mineral matrix depositor cells. We suggest that DEX may be omitted from culture media supplementation for SHED osteogenic differentiation in vitro studies.
Collapse
Affiliation(s)
- Mariane Beatriz Sordi
- Center for Research on Dental Implants, Federal University of Santa Catarina, Delfino Conti Street, Florianópolis, 88040-900, Brazil
- Laboratory of Applied Virology, Federal University of Santa Catarina, Henrique da Silva Fontes Avenue, Florianópolis, 88040-900, Brazil
| | - Raissa Borges Curtarelli
- Center for Research on Dental Implants, Federal University of Santa Catarina, Delfino Conti Street, Florianópolis, 88040-900, Brazil
- Laboratory of Applied Virology, Federal University of Santa Catarina, Henrique da Silva Fontes Avenue, Florianópolis, 88040-900, Brazil
| | - Izabella Thaís da Silva
- Laboratory of Applied Virology, Federal University of Santa Catarina, Henrique da Silva Fontes Avenue, Florianópolis, 88040-900, Brazil
- Department of Pharmaceutics Science, Federal University of Santa Catarina, Delfino Conti Street, Florianópolis, 88040-900, Brazil
| | - Gislaine Fongaro
- Laboratory of Applied Virology, Federal University of Santa Catarina, Henrique da Silva Fontes Avenue, Florianópolis, 88040-900, Brazil
- Department of Microbiology, Immunology, and Parasitology, Federal University of Santa Catarina, Henrique da Silva Fontes Avenue, Florianópolis, 88040-900, Brazil
| | - Cesar Augusto Magalhães Benfatti
- Center for Research on Dental Implants, Federal University of Santa Catarina, Delfino Conti Street, Florianópolis, 88040-900, Brazil
- Department of Dentistry, Federal University of Santa Catarina, Delfino Conti Street, Florianópolis, 88040-900, Brazil
| | - Ricardo de Souza Magini
- Center for Research on Dental Implants, Federal University of Santa Catarina, Delfino Conti Street, Florianópolis, 88040-900, Brazil
- Department of Dentistry, Federal University of Santa Catarina, Delfino Conti Street, Florianópolis, 88040-900, Brazil
| | - Ariadne Cristiane Cabral da Cruz
- Center for Research on Dental Implants, Federal University of Santa Catarina, Delfino Conti Street, Florianópolis, 88040-900, Brazil.
- Laboratory of Applied Virology, Federal University of Santa Catarina, Henrique da Silva Fontes Avenue, Florianópolis, 88040-900, Brazil.
- Department of Dentistry, Federal University of Santa Catarina, Delfino Conti Street, Florianópolis, 88040-900, Brazil.
| |
Collapse
|
14
|
Chakraborty S, Sinha S, Sengupta A. Emerging trends in chromatin remodeler plasticity in mesenchymal stromal cell function. FASEB J 2020; 35:e21234. [PMID: 33337557 DOI: 10.1096/fj.202002232r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022]
Abstract
Emerging evidences highlight importance of epigenetic regulation and their integration with transcriptional and cell signaling machinery in determining tissue resident adult pluripotent mesenchymal stem/stromal cell (MSC) activity, lineage commitment, and multicellular development. Histone modifying enzymes and large multi-subunit chromatin remodeling complexes and their cell type-specific plasticity remain the central defining features of gene regulation and establishment of tissue identity. Modulation of transcription factor expression gradient ex vivo and concomitant flexibility of higher order chromatin architecture in response to signaling cues are exciting approaches to regulate MSC activity and tissue rejuvenation. Being an important constituent of the adult bone marrow microenvironment/niche, pathophysiological perturbation in MSC homeostasis also causes impaired hematopoietic stem/progenitor cell function in a non-cell autonomous mechanism. In addition, pluripotent MSCs can function as immune regulatory cells, and they reside at the crossroad of innate and adaptive immune response pathways. Research in the past few years suggest that MSCs/stromal fibroblasts significantly contribute to the establishment of immunosuppressive microenvironment in shaping antitumor immunity. Therefore, it is important to understand mesenchymal stromal epigenome and transcriptional regulation to leverage its applications in regenerative medicine, epigenetic memory-guided trained immunity, immune-metabolic rewiring, and precision immune reprogramming. In this review, we highlight the latest developments and prospects in chromatin biology in determining MSC function in the context of lineage commitment and immunomodulation.
Collapse
Affiliation(s)
- Sayan Chakraborty
- Stem Cell & Leukemia Laboratory, Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,Translational Research Unit of Excellence (TRUE), Kolkata, India
| | - Sayantani Sinha
- Stem Cell & Leukemia Laboratory, Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,Translational Research Unit of Excellence (TRUE), Kolkata, India
| | - Amitava Sengupta
- Stem Cell & Leukemia Laboratory, Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.,Translational Research Unit of Excellence (TRUE), Kolkata, India
| |
Collapse
|
15
|
Ha N, Choi YI, Jung N, Song JY, Bae DK, Kim MC, Lee YJ, Song H, Kwak G, Jeong S, Park S, Nam SH, Jung S, Choi B. A novel histone deacetylase 6 inhibitor improves myelination of Schwann cells in a model of Charcot-Marie-Tooth disease type 1A. Br J Pharmacol 2020; 177:5096-5113. [PMID: 33460073 PMCID: PMC7589015 DOI: 10.1111/bph.15231] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 07/22/2020] [Accepted: 07/25/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Charcot-Marie-Tooth (CMT) disease is the most common hereditary peripheral neuropathy. CMT type 1A (CMT1A) accounts for approximately 50% of CMT patients and is linked to PMP22 gene duplication. Histone deacetylase-6 (HDAC6) has pleiotropic effects, such as regulating lipid homeostasis and cellular stress. Although HDAC6 has been regarded as a promising drug target for neurodegenerative diseases, its inhibition has not yet been tested in CMT1A. Here we have tested the therapeutic potential of CKD-504, a clinical stage HDAC6 inhibitor, in a mouse model of CMT1A EXPERIMENTAL APPROACH: The potency and selectivity of CKD-504 was evaluated, using a HDAC enzyme panel assay and western blots. The therapeutic potential of CKD-504 was evaluated using behavioural testing and electrophysiological assessments in the C22 mouse model of CMT1A. PMP22 protein expression and aggregation were analysed in mesenchymal stem cell-derived Schwann cells from CMT1A patients and sciatic nerves from C22 mice. KEY RESULTS The HDAC6 inhibitor, CKD-504, modulated molecular chaperon proteins such as HSP90 and HSP70, which are involved in the folding/refolding of proteins such as PMP22. CKD-504 treatment restored myelination in both mesenchymal stem cell-derived Schwann cells from CMT1A patients and sciatic nerves of C22 mice and improved the axonal integrity of the sciatic nerve, leading to behavioural, electrophysiological, and histological improvements in C22 mice. CONCLUSION AND IMPLICATIONS A novel HDAC6 inhibitor, CKD-504, has potent therapeutic efficacy for CMT1A.
Collapse
Affiliation(s)
- Nina Ha
- Department of Health Sciences and Technology, SAIHSTSungkyunkwan UniversitySeoulRepublic of Korea
- CKD Research InstituteYonginRepublic of Korea
| | | | - Namhee Jung
- Department of Biochemistry, College of MedicineEwha Womans UniversitySeoulRepublic of Korea
| | | | | | | | | | | | - Geon Kwak
- Department of Health Sciences and Technology, SAIHSTSungkyunkwan UniversitySeoulRepublic of Korea
| | - Soyeon Jeong
- Department of Biochemistry, College of MedicineEwha Womans UniversitySeoulRepublic of Korea
| | - Saeyoung Park
- Department of Biochemistry, College of MedicineEwha Womans UniversitySeoulRepublic of Korea
| | - Soo Hyun Nam
- Department of Neurology, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Sung‐Chul Jung
- Department of Biochemistry, College of MedicineEwha Womans UniversitySeoulRepublic of Korea
| | - Byung‐Ok Choi
- Department of Health Sciences and Technology, SAIHSTSungkyunkwan UniversitySeoulRepublic of Korea
- Department of Neurology, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulRepublic of Korea
| |
Collapse
|
16
|
LncRNA DANCR and miR-320a suppressed osteogenic differentiation in osteoporosis by directly inhibiting the Wnt/β-catenin signaling pathway. Exp Mol Med 2020; 52:1310-1325. [PMID: 32778797 PMCID: PMC8080634 DOI: 10.1038/s12276-020-0475-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/26/2020] [Accepted: 06/10/2020] [Indexed: 12/19/2022] Open
Abstract
Our study aimed to determine how lncRNA DANCR, miR-320a, and CTNNB1 interact with each other and regulate osteogenic differentiation in osteoporosis. qRT-PCR and western blotting were performed to determine the expression of DANCR, miR-320a, CTNNB1, and the osteoporosis- or Wnt/β-catenin pathway-related markers T-cell factor 1 (TCF-1), runt-related transcription factor 2 (RUNX2), alkaline phosphatase (ALP), osteocalcin (OCN), and osteopontin (OPN). Interactions between CTNNB1, DANCR, and miR-320a were predicted by bioinformatics approaches and validated using a luciferase assay. Osteoblastic phenotypes were evaluated by ALP staining, ALP activity assay and Alizarin Red staining. The bilateral ovariectomy method was used to establish an in vivo osteoporosis model. Bone morphological changes were examined using hematoxylin and eosin (H&E) and Alcian Blue staining. The expression levels of DANCR and miR-320a in BMSCs derived from osteoporosis patients were upregulated, whereas CTNNB1 expression was downregulated compared with that in healthy controls. Importantly, we demonstrated that miR-320a and DANCR acted independently from each other and both inhibited CTNNB1 expression, whereas the inhibitory effect was additive when miR-320a and DANCR were cooverexpressed. Moreover, we found that DANCR overexpression largely abrogated the effect of the miR-320a inhibitor on CTNNB1 expression and the Wnt/β-catenin signaling pathway in BMSCs during osteogenic differentiation. We further confirmed the results above in BMSCs derived from an osteoporosis animal model. Taken together, our findings revealed that DANCR and miR-320a regulated the Wnt/β-catenin signaling pathway during osteogenic differentiation in osteoporosis through CTNNB1 inhibition. Our results highlight the potential value of DANCR and miR-320a as promising therapeutic targets for osteoporosis treatment. Two non-coding RNAs are potential targets for reducing bone loss in post-menopausal osteoporosis. Bones are constantly being remodeled; when resorption outpaces generation of new bone, bones are weakened, causing osteoporosis and leading to decreased quality of life and injuries. Although treatments exist, they often have undesirable side effects, and new treatments are needed. The molecular basis of the changes that accompany osteoporosis are poorly understood. Da Zhong at the Xiangya Hospital of Central South University in Changsha, China, and co-workers investigated how two non-coding RNAs, small molecules that regulate gene expression, are involved in the progression of post-menopausal osteoporosis. They found that levels of both molecules are increased in osteoporosis, and that silencing them increases building of new bone, key to maintaining bone strength. These results illuminate a potential new direction in treatments for osteoporosis.
Collapse
|
17
|
Wang N, Wang H, Chen J, Wang F, Wang S, Zhou Q, Ying J, Huang S, Wang P, Yuan F. ACY‑1215, a HDAC6 inhibitor, decreases the dexamethasone‑induced suppression of osteogenesis in MC3T3‑E1 cells. Mol Med Rep 2020; 22:2451-2459. [PMID: 32705192 PMCID: PMC7411391 DOI: 10.3892/mmr.2020.11319] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoid-induced osteoporosis is the commonest form of drug-induced osteoporosis. Histone deacetylase 6 (HDAC6) is involved in the differentiation from mesenchymal stem cells to osteoblasts. However, the role of ricolinostat (ACY-1215, HDAC6 inhibitor) in the dexamethasone (Dex)-induced proliferation and differentiation of preosteoblasts remains to be elucidated. The protein expression and mRNA expression levels of HDAC6, osteopontin (OPN), runt-related transcription factor 2 (Runx2), osterix (Osx), collagen I (COL1A1) and glucocorticoid receptor (GR) in MC3T3-E1 cells were analyzed by western blot analysis and reverse transcription-quantitative PCR analysis. The cell viability was detected by CCK-8 assay. The alkaline phosphatase (ALP) activity and capacity of mineralization was determined by ALP assay kit and alizarin red staining. HDAC6 expression was increased in patient serum and Dex-induced MC3T3-E1 cells at a certain concentration range; 1 µM Dex was selected for further experimentation. Cell viability was decreased after Dex induction and restored following ACY-1215 treatment. The ALP activity and capability for mineralization was decreased when MC3T3-E1 cells were induced by 1 µM Dex and was gradually improved by the treatment of ACY-1215 at 1, 5 and 10 mM. The expression of OPN, Runx2, Osx and COL1A1 was similar, with the changes of capability for mineralization. Furthermore, GR expression was increased in Dex-induced MC3T3-E1 cells. ACY-1215 promoted the GR expression in MC3T3-E1 cells from 1–5 mM while GR receptor expression was increased with 10 mM ACY-1215 treatment. In conclusion, ACY-1215 reversed the Dex-induced suppression of proliferation and differentiation of MC3T3-E1 cells.
Collapse
Affiliation(s)
- Na Wang
- Department of Endocrinology, Ningbo Number 6 Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Hua Wang
- Department of Radiology, Ningbo Number 6 Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Jianming Chen
- Department of Orthopaedics, Ningbo Number 6 Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Fubin Wang
- Department of Inspection, Ningbo Number 6 Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Shuaiyi Wang
- Department of Orthopaedics, Ningbo Number 6 Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Qiang Zhou
- Department of Endocrinology, The First Hospital of Jiaxing (The Affiliated Hospital of Jiaxing University), Jiaxing, Zhejiang 314001, P.R. China
| | - Jichong Ying
- Department of Orthopaedics, Ningbo Number 6 Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Shanzhao Huang
- Department of Osteopeniology, Ningbo Number 6 Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Pu Wang
- Department of Cardiology, Ningbo Number 6 Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Fangfang Yuan
- Department of Rheumatology, Ningbo Number 6 Hospital, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
18
|
Claeys L, Bravenboer N, Eekhoff EMW, Micha D. Human Fibroblasts as a Model for the Study of Bone Disorders. Front Endocrinol (Lausanne) 2020; 11:394. [PMID: 32636804 PMCID: PMC7318867 DOI: 10.3389/fendo.2020.00394] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 05/18/2020] [Indexed: 01/03/2023] Open
Abstract
Bone tissue degeneration is an urgent clinical issue, making it a subject of intensive research. Chronic skeletal disease forms can be prevalent, such as the age-related osteoporosis, or rare, in the form of monogenetic bone disorders. A barrier in the understanding of the underlying pathological process is the lack of accessibility to relevant material. For this reason, cells of non-bone tissue are emerging as a suitable alternative for models of bone biology. Fibroblasts are highly suitable for this application; they populate accessible anatomical locations, such as the skin tissue. Reports suggesting their utility in preclinical models for the study of skeletal diseases are increasingly becoming available. The majority of these are based on the generation of an intermediate stem cell type, the induced pluripotent stem cells, which are subsequently directed to the osteogenic cell lineage. This intermediate stage is circumvented in transdifferentiation, the process regulating the direct conversion of fibroblasts to osteogenic cells, which is currently not well-explored. With this mini review, we aimed to give an overview of existing osteogenic transdifferentiation models and to inform about their applications in bone biology models.
Collapse
Affiliation(s)
- Lauria Claeys
- Department of Clinical Genetics, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Elisabeth M. W. Eekhoff
- Department of Internal Medicine Section Endocrinology, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Dimitra Micha
- Department of Clinical Genetics, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
19
|
Kershaw S, Morgan DJ, Boyd J, Spiller DG, Kitchen G, Zindy E, Iqbal M, Rattray M, Sanderson CM, Brass A, Jorgensen C, Hussell T, Matthews LC, Ray DW. Glucocorticoids rapidly inhibit cell migration through a novel, non-transcriptional HDAC6 pathway. J Cell Sci 2020; 133:jcs242842. [PMID: 32381682 PMCID: PMC7295589 DOI: 10.1242/jcs.242842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoids (GCs) act through the glucocorticoid receptor (GR, also known as NR3C1) to regulate immunity, energy metabolism and tissue repair. Upon ligand binding, activated GR mediates cellular effects by regulating gene expression, but some GR effects can occur rapidly without new transcription. Here, we show that GCs rapidly inhibit cell migration, in response to both GR agonist and antagonist ligand binding. The inhibitory effect on migration is prevented by GR knockdown with siRNA, confirming GR specificity, but not by actinomycin D treatment, suggesting a non-transcriptional mechanism. We identified a rapid onset increase in microtubule polymerisation following GC treatment, identifying cytoskeletal stabilisation as the likely mechanism of action. HDAC6 overexpression, but not knockdown of αTAT1, rescued the GC effect, implicating HDAC6 as the GR effector. Consistent with this hypothesis, ligand-dependent cytoplasmic interaction between GR and HDAC6 was demonstrated by quantitative imaging. Taken together, we propose that activated GR inhibits HDAC6 function, and thereby increases the stability of the microtubule network to reduce cell motility. We therefore report a novel, non-transcriptional mechanism whereby GCs impair cell motility through inhibition of HDAC6 and rapid reorganization of the cell architecture.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Stephen Kershaw
- Systems Oncology, Cancer Research UK Manchester Institute, Manchester, SK10 4TG, UK
| | - David J Morgan
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, M13 9PT, UK
- Lydia Becker Institute of Immunology and Inflammation University of Manchester, Manchester, M13 9PT, UK
| | - James Boyd
- Division of Cellular and Molecular Physiology, University of Liverpool, Liverpool, L69 3BX, UK
| | - David G Spiller
- Platform Sciences, Enabling Technologies, and Infrastructure, University of Manchester, Manchester, M13 9PT, UK
| | - Gareth Kitchen
- Division of Diabetes, Endocrinology, and Gastroenterology, University of Manchester, Manchester, M13 9PT, UK
| | - Egor Zindy
- Division of Informatics, Imaging, and Data Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Mudassar Iqbal
- Division of Informatics, Imaging, and Data Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Magnus Rattray
- Division of Informatics, Imaging, and Data Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Christopher M Sanderson
- Division of Cellular and Molecular Physiology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Andrew Brass
- Division of Informatics, Imaging, and Data Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Claus Jorgensen
- Systems Oncology, Cancer Research UK Manchester Institute, Manchester, SK10 4TG, UK
| | - Tracy Hussell
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, M13 9PT, UK
- Lydia Becker Institute of Immunology and Inflammation University of Manchester, Manchester, M13 9PT, UK
| | - Laura C Matthews
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - David W Ray
- Division of Diabetes, Endocrinology, and Gastroenterology, University of Manchester, Manchester, M13 9PT, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, OX3 7LE, and NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| |
Collapse
|
20
|
The Role of DNMT and HDACs in the Fetal Programming of Hypertension by Glucocorticoids. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5751768. [PMID: 32318239 PMCID: PMC7149440 DOI: 10.1155/2020/5751768] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/07/2020] [Indexed: 12/13/2022]
Abstract
The causes of hypertension are complex and involve both genetic and environmental factors. Environment changes during fetal development have been linked to adult diseases including hypertension. Studies show that timed in utero exposure to the synthetic glucocorticoid (GC) dexamethasone (Dex) results in the development of hypertension in adult rats. Evidence suggests that in utero stress can alter patterns of gene expression, possibly a result of alterations in the topology of the genome by epigenetic markers such as DNA methyltransferases (DNMTs) and histone deacetylases (HDACs). The objective of this study was to determine the effects of epigenetic regulators in the fetal programming and the development of adult hypertension. Specifically, this research examined the effects of the HDAC inhibitor valproic acid (VPA) and the DNMT inhibitor 5-aza-2′-deoxycytidine (5aza2DC) on blood pressure (BP) and gene expression in prenatal Dex-programmed rats. Data suggest that both VPA and 5aza2DC attenuated the Dex-mediated development of hypertension and restored BP to control levels. Epigenetic DNMT inhibition (DNMTi) or HDAC inhibition (HDACi) also successfully attenuated elevations in the majority of altered catecholamine (CA) enzyme expression, phenylethanolamine N-methyltransferase (PNMT) protein, and elevated epinephrine (Epi) levels in males. Although females responded to HDACi similar to males, DNMTi drove increased glucocorticoid receptor (GR) and PNMT expression and elevations in circulating Epi in females despite showing normotensive BP.
Collapse
|
21
|
Ren J, Huang D, Li R, Wang W, Zhou C. Control of mesenchymal stem cell biology by histone modifications. Cell Biosci 2020; 10:11. [PMID: 32025282 PMCID: PMC6996187 DOI: 10.1186/s13578-020-0378-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 01/24/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are considered the most promising seed cells for regenerative medicine because of their considerable therapeutic properties and accessibility. Fine-tuning of cell biological processes, including differentiation and senescence, is essential for achievement of the expected regenerative efficacy. Researchers have recently made great advances in understanding the spatiotemporal gene expression dynamics that occur during osteogenic, adipogenic and chondrogenic differentiation of MSCs and the intrinsic and environmental factors that affect these processes. In this context, histone modifications have been intensively studied in recent years and have already been indicated to play significant and universal roles in MSC fate determination and differentiation. In this review, we summarize recent discoveries regarding the effects of histone modifications on MSC biology. Moreover, we also provide our insights and perspectives for future applications.
Collapse
Affiliation(s)
- Jianhan Ren
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou, 510055 China
| | - Delan Huang
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou, 510055 China
| | - Runze Li
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou, 510055 China
| | - Weicai Wang
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou, 510055 China
| | - Chen Zhou
- Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Guangzhou, 510055 China
| |
Collapse
|
22
|
Lin Z, Li Z, Li EN, Li X, Del Duke CJ, Shen H, Hao T, O'Donnell B, Bunnell BA, Goodman SB, Alexander PG, Tuan RS, Lin H. Osteochondral Tissue Chip Derived From iPSCs: Modeling OA Pathologies and Testing Drugs. Front Bioeng Biotechnol 2019; 7:411. [PMID: 31921815 PMCID: PMC6930794 DOI: 10.3389/fbioe.2019.00411] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/27/2019] [Indexed: 01/17/2023] Open
Abstract
Osteoarthritis (OA) is a chronic disease mainly characterized by degenerative changes in cartilage, but other joint elements such as bone are also affected. To date, there are no disease-modifying OA drugs (DMOADs), owing in part to a deficiency of current models in simulating OA pathologies and etiologies in humans. In this study, we aimed to develop microphysiological osteochondral (OC) tissue chips derived from human induced pluripotent stem cells (iPSCs) to model the pathologies of OA. We first induced iPSCs into mesenchymal progenitor cells (iMPCs) and optimized the chondro- and osteo-inductive conditions for iMPCs. Then iMPCs were encapsulated into photocrosslinked gelatin scaffolds and cultured within a dual-flow bioreactor, in which the top stream was chondrogenic medium and the bottom stream was osteogenic medium. After 28 days of differentiation, OC tissue chips were successfully generated and phenotypes were confirmed by real time RT-PCR and histology. To create an OA model, interleukin-1β (IL-1β) was used to challenge the cartilage component for 7 days. While under control conditions, the bone tissue promoted chondrogenesis and suppressed chondrocyte terminal differentiation of the overlying chondral tissue. Under conditions modeling OA, the bone tissue accelerated the degradation of chondral tissue which is likely via the production of catabolic and inflammatory cytokines. These findings suggest active functional crosstalk between the bone and cartilage tissue components in the OC tissue chip under both normal and pathologic conditions. Finally, a selective COX-2 inhibitor commonly prescribed drug for OA, Celecoxib, was shown to downregulate the expression of catabolic and proinflammatory cytokines in the OA model, demonstrating the utility of the OC tissue chip model for drug screening. In summary, the iPSC-derived OC tissue chip developed in this study represents a high-throughput platform applicable for modeling OA and for the screening and testing of candidate DMOADs.
Collapse
Affiliation(s)
- Zixuan Lin
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhong Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Eileen N. Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
| | - Xinyu Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
| | - Colin J. Del Duke
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - He Shen
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Tingjun Hao
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
| | - Benjamen O'Donnell
- Department of Pharmacology, Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA, United States
| | - Bruce A. Bunnell
- Department of Pharmacology, Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA, United States
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery and Bioengineering, Stanford University, Stanford, CA, United States
| | - Peter G. Alexander
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Rocky S. Tuan
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Hang Lin
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
23
|
Fahimipour F, Dashtimoghadam E, Mahdi Hasani-Sadrabadi M, Vargas J, Vashaee D, Lobner DC, Jafarzadeh Kashi TS, Ghasemzadeh B, Tayebi L. Enhancing cell seeding and osteogenesis of MSCs on 3D printed scaffolds through injectable BMP2 immobilized ECM-Mimetic gel. Dent Mater 2019; 35:990-1006. [PMID: 31027908 PMCID: PMC7193177 DOI: 10.1016/j.dental.2019.04.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Design of bioactive scaffolds with osteogenic capacity is a central challenge in cell-based patient-specific bone tissue engineering. Efficient and spatially uniform seeding of (stem) cells onto such constructs is vital to attain functional tissues. Herein we developed heparin functionalized collagen gels supported by 3D printed bioceramic scaffolds, as bone extracellular matrix (ECM)-mimetic matrices. These matrices were designed to enhance cell seeding efficiency of mesenchymal stem cells (MSCs) as well as improve their osteogenic differentiation through immobilized bone morphogenic protein 2 (BMP2) to be used for personalized bone regeneration. METHODS A 3D gel based on heparin-conjugated collagen matrix capable of immobilizing recombinant human bone morphogenic protein 2 (BMP2) was synthesized. Isolated dental pulp Mesenchymal stem cells (MSCs) were then encapsulated into the bone ECM microenvironment to efficiently and uniformly seed a bioactive ceramic-based scaffold fabricated using additive manufacturing technique. The designed 3D cell-laden constructs were comprehensively investigated trough in vitro assays and in vivo study. RESULTS In-depth rheological characterizations of heparin-conjugated collagen gel revealed that elasticity of the matrix is significantly improved compared with freely incorporated heparin. Investigation of the MSCs laden collagen-heparin hydrogels revealed their capability to provide spatiotemporal bioavailability of BMP2 while suppressing the matrix contraction over time. The in vivo histology and real-time polymerase chain reaction (qPCR) analysis showed that the designed construct supported the osteogenic differentiation of MSCs and induced the ectopic bone formation in rat model. SIGNIFICANCE The presented hybrid constructs combine bone ECM chemical cues with mechanical function providing an ideal 3D microenvironment for patient-specific bone tissue engineering and cell therapy applications. The implemented methodology in design of ECM-mimetic 3D matrix capable of immobilizing BMP2 to improve seeding efficiency of customized scaffolds can be exploited for other bioactive molecules.
Collapse
Affiliation(s)
- Farahnaz Fahimipour
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA; Dental Biomaterials Department, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Mahdi Hasani-Sadrabadi
- Parker H. Petit Institute for Bioengineering and Bioscience, G.W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prothodontics, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Jessica Vargas
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Daryoosh Vashaee
- Electrical and Computer Engineering Department, North Carolina State University, Raleigh, NC 27606, USA
| | - Douglas C Lobner
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - Tahereh S Jafarzadeh Kashi
- Dental Biomaterials Department, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnam Ghasemzadeh
- Department of Biomedical Sciences, Integrative Neuroscience Research Center, Marquette University, Milwaukee, WI 53201, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA.
| |
Collapse
|
24
|
Ehnert S, Aspera-Werz RH, Ruoß M, Dooley S, Hengstler JG, Nadalin S, Relja B, Badke A, Nussler AK. Hepatic Osteodystrophy-Molecular Mechanisms Proposed to Favor Its Development. Int J Mol Sci 2019; 20:2555. [PMID: 31137669 PMCID: PMC6566554 DOI: 10.3390/ijms20102555] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/14/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023] Open
Abstract
Almost all patients with chronic liver diseases (CLD) show altered bone metabolism. Depending on the etiology, this manifests in a severe osteoporosis in up to 75% of the affected patients. Due to high prevalence, the generic term hepatic osteodystrophy (HOD) evolved, describing altered bone metabolism, decreased bone mineral density, and deterioration of bone structure in patients with CLD. Once developed, HOD is difficult to treat and increases the risk of fragility fractures. Existing fractures affect the quality of life and, more importantly, long-term prognosis of these patients, which presents with increased mortality. Thus, special care is required to support the healing process. However, for early diagnosis (reduce fracture risk) and development of adequate treatment strategies (support healing of existing fractures), it is essential to understand the underlying mechanisms that link disturbed liver function with this bone phenotype. In the present review, we summarize proposed molecular mechanisms favoring the development of HOD and compromising the healing of associated fractures, including alterations in vitamin D metabolism and action, disbalances in transforming growth factor beta (TGF-β) and bone morphogenetic protein (BMP) signaling with histone deacetylases (HDACs) as secondary regulators, as well as alterations in the receptor activator of nuclear factor kappa B ligand (RANKL)-osteoprotegerin (OPG) system mediated by sclerostin. Based on these mechanisms, we give an overview on the limitations of early diagnosis of HOD with established serum markers.
Collapse
Affiliation(s)
- Sabrina Ehnert
- Siegfried Weller Research Institute, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Romina H Aspera-Werz
- Siegfried Weller Research Institute, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Marc Ruoß
- Siegfried Weller Research Institute, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Steven Dooley
- Department of Medicine II, Molecular Hepatology, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany.
| | - Jan G Hengstler
- IfADo-Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, 44139 Dortmund, Germany.
| | - Silvio Nadalin
- Department of General, Visceral and Transplant Surgery, University Hospital Tuebingen, 72076 Tuebingen, Germany.
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany.
| | - Andreas Badke
- Siegfried Weller Research Institute, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Andreas K Nussler
- Siegfried Weller Research Institute, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| |
Collapse
|
25
|
Mohamed Hassan Y, Massa L, Caviglia C, Sylvest Keller S. Electrochemical Monitoring of Saos-2 Cell Differentiation on Pyrolytic Carbon Electrodes. ELECTROANAL 2018. [DOI: 10.1002/elan.201800429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Yasmin Mohamed Hassan
- Department of Micro- and Nanotechnology; Technical University of Denmark; 2800 Kongens Lyngby Denmark
| | - Leonardo Massa
- Department of Micro- and Nanotechnology; Technical University of Denmark; 2800 Kongens Lyngby Denmark
| | - Claudia Caviglia
- Department of Micro- and Nanotechnology; Technical University of Denmark; 2800 Kongens Lyngby Denmark
| | - Stephan Sylvest Keller
- Department of Micro- and Nanotechnology; Technical University of Denmark; 2800 Kongens Lyngby Denmark
| |
Collapse
|
26
|
Zhao M, Li P, Xu H, Pan Q, Zeng R, Ma X, Li Z, Lin H. Dexamethasone-Activated MSCs Release MVs for Stimulating Osteogenic Response. Stem Cells Int 2018; 2018:7231739. [PMID: 29760734 PMCID: PMC5926524 DOI: 10.1155/2018/7231739] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 02/22/2018] [Indexed: 02/07/2023] Open
Abstract
The extracellular microvesicles (MVs) are attracting much attention because they are found to be the key paracrine mediator participating in tissue regeneration. Dexamethasone (DXM) is widely accepted as an important regulator in tailoring the differentiation potential of mesenchymal stem cells (MSCs). However, the effect of DXM on the paracrine signaling of MSCs remains unknown. To this point, we aimed to explore the role of DXM in regulating the paracrine activity of MSCs through evaluating the release and function of MSC-MVs, based on their physicochemical characteristics and support on osteogenic response. Results showed that DXM had no evident impact on the release of MSC-MVs but played a pivotal role in regulating the function of MSC-MVs. MVs obtained from the DXM-stimulated MSCs (DXM-MVs) increased MC3T3 cell proliferation and migration and upregulated Runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP), and osteopontin (OPN) expression. The repair efficiency of DXM-MVs for femur defects was further investigated in an established rat model. It was found that DXM-MVs accelerated the healing process of bone formation in the defect area. Thus, we conclude that using DXM as stimuli to obtain functional MSCs-MVs could become a valuable tool for promoting bone regeneration.
Collapse
Affiliation(s)
- Mingyan Zhao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Peng Li
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Haijia Xu
- Department of Orthopaedics, Tongren Hospital of Wuhan University, Wuhan 430060, China
| | - Qunwen Pan
- Department of Surgery, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Rong Zeng
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Xiaotang Ma
- Department of Surgery, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Zhanghua Li
- Department of Orthopaedics, Tongren Hospital of Wuhan University, Wuhan 430060, China
| | - Hao Lin
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| |
Collapse
|
27
|
Zhou D, Chen YX, Yin JH, Tao SC, Guo SC, Wei ZY, Feng Y, Zhang CQ. Valproic acid prevents glucocorticoid‑induced osteonecrosis of the femoral head of rats. Int J Mol Med 2018; 41:3433-3447. [PMID: 29512684 PMCID: PMC5881807 DOI: 10.3892/ijmm.2018.3534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 02/28/2018] [Indexed: 01/26/2023] Open
Abstract
Glucocorticoids (GCs) are the most common cause of atraumatic osteonecrosis of the femoral head (ONFH) because their effect compromises the osteogenic capability of bone marrow-derived mesenchymal stem cells (BMSCs). Valproic acid (VPA) is a widely used anti-epileptic and anticonvulsant drug. Previous studies have reported that VPA promotes osteogenic differentiation of MSCs in vitro and osteogenesis in vivo as a histone deacetylase (HDAC) inhibitor. The purpose of the present study was to investigate the efficacy of VPA as a precautionary treatment of ONFH after GC treatment in rats. In vitro, the effect of VPA, dexamethasone or a combination treatment of the two on the proliferation and osteogenic differentiation of human BMSCs was assessed using a Cell Counting Kit-8 and apoptosis assays, and by measuring the expression of proteins associated with osteogenesis. In vivo, a GC-induced ONFH model was established in rats and VPA was added during GC treatment to investigate the preventive effect of VPA against ONFH. Rat BMSCs were also extracted to investigate the osteogenic capacity. The results of micro-computed tomography scanning, angiography of the femoral head and histological and immunohistochemical analyses indicated that 11 of 15 rats induced with methylprednisolone (MP) presented with ONFH, while only 2 of 15 rats treated with a combination of MP and VPA developed ONFH. VPA produced beneficial effects on subchondral bone trabeculae in the femoral head with significant preservation of bone volume and blood supply, as well as improved osteogenic capability of BMSCs compared with those in rats treated with GC alone. In conclusion, VPA attenuated the inhibitory effect of GC on BMSC proliferation and osteogenesis by inhibiting apoptosis and elevating the expression of proteins associated with osteogenesis, which may contribute to the prevention of GC-induced ONFH in rats.
Collapse
Affiliation(s)
- Ding Zhou
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yi-Xuan Chen
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Jun-Hui Yin
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Shi-Cong Tao
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Shang-Chun Guo
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Zhan-Ying Wei
- Metabolic Bone Disease and Genetic Research Unit, Division of Osteoporosis and Bone Disease, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yong Feng
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Chang-Qing Zhang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
28
|
Chen SM, Peng YJ, Wang CC, Su SL, Salter DM, Lee HS. Dexamethasone Down-regulates Osteocalcin in Bone Cells through Leptin Pathway. Int J Med Sci 2018; 15:507-516. [PMID: 29559840 PMCID: PMC5859774 DOI: 10.7150/ijms.21881] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 01/05/2018] [Indexed: 01/26/2023] Open
Abstract
Glucocorticoid therapy, especially at higher doses, is associated with significant adverse side effects including osteoporosis. Leptin, secreted from adipose tissue, has diverse effects on bone tissue regulation. As glucocorticoids stimulate leptin synthesis and secretion directly in adipose tissue we hypothesised that dexamethasone (DEX) induced osteoporosis may, in part, be mediated by an osteoblast dependent leptin-leptin receptor pathway. Human bone cells expressed leptin and leptin receptors (Ob-Ra and Ob-Rb). DEX increased leptin, Ob-Ra and Ob-Rb expression in a dose-dependent manner while decreasing expression of osteocalcin. In the presence of leptin, Cbfa1 and osteonectin expression showed no significant change, whereas osteocalcin expression was decreased. Recombinant human quadruple antagonist leptin suppressed DEX-induced osteocalcin downregulation. The signaling pathway involved up-regulation of JAK2. In conclusion, upregulation of leptin and Ob-Rb in human bone cells by DEX is associated with down-regulation of osteocalcin expression. The down regulation of osteocalcin by DEX was partially through a leptin autocrine/paracrine loop. Adverse effects of DEX on the skeleton may be modified by targeting leptin signaling pathways.
Collapse
Affiliation(s)
- Shu-Mei Chen
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC.,Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan, ROC
| | - Yi-Jen Peng
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chih-Chien Wang
- Department of Orthopedics, Taipei Medical University Hospital, Taipei, Taiwan, ROC
| | - Sui-Lung Su
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Donald M Salter
- Centre for Genomic and Molecular Medicine, IGMM, University of Edinburgh, Edinburgh, UK
| | - Herng-Sheng Lee
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| |
Collapse
|
29
|
Ren H, Shen G, Tang J, Qiu T, Zhang Z, Zhao W, Yu X, Huang J, Liang D, Yao Z, Yang Z, Jiang X. Promotion effect of extracts from plastrum testudinis on alendronate against glucocorticoid-induced osteoporosis in rat spine. Sci Rep 2017; 7:10617. [PMID: 28878388 PMCID: PMC5587701 DOI: 10.1038/s41598-017-10614-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022] Open
Abstract
Alendronate (ALN) is a key therapeutic used to treat glucocorticoid-induced osteoporosis (GIOP), but may induce severe side effects. We showed earlier that plastrum testudinis extracts (PTE) prevented and treated GIOP in vivo. However, clinically, PTE is seldom used alone. Herein, we reveal the synergistic effect of ALN and PTE can treat GIOP of the rat spine and define the mechanism. Sprague-Dawley rats were randomly assigned to four groups: a vehicle group, a GIOP group, an ALN group, and an ALN+PTE group. Each group was further divided into two experimental phases, including dexamethasone (DXM) intervention and withdrawal. Bone mass, microarchitecture, biomechanics, bone-turnover markers, and histomorphology were evaluated. The mRNA and protein expression levels of CTSK and Runx2 were detemined. We found that ALN+PTE improved bone quantity and quality, bone strength, bone turnover; and mitigated histological damage during glucocorticoid intervention and withdrawal. The therapeutic effect was better than that afforded by ALN alone. ALN+PTE reduced CTSK protein expression, promoted Runx2 mRNA and protein expression to varying extents, and more strongly inhibited bone resorption than did ALN alone. Overall, the synergistic effect mediated by ALN+PTE reversed GIOP during DXM intervention and withdrawal via affecting CTSK and Runx2 expression at mRNA and protein levels.
Collapse
Affiliation(s)
- Hui Ren
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou, Guangzhou, 510405, China
| | - Gengyang Shen
- Guangzhou University of Chinese Medicine Guangzhou , Guangzhou, 510405, China
| | - Jingjing Tang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou, Guangzhou, 510405, China
| | - Ting Qiu
- Guangzhou University of Chinese Medicine Guangzhou , Guangzhou, 510405, China
| | - Zhida Zhang
- Guangzhou University of Chinese Medicine Guangzhou , Guangzhou, 510405, China
| | - Wenhua Zhao
- Guangzhou University of Chinese Medicine Guangzhou , Guangzhou, 510405, China
| | - Xiang Yu
- Guangzhou University of Chinese Medicine Guangzhou , Guangzhou, 510405, China
| | - Jinjing Huang
- Guangzhou University of Chinese Medicine Guangzhou , Guangzhou, 510405, China
| | - De Liang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou, Guangzhou, 510405, China
| | - Zhensong Yao
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou, Guangzhou, 510405, China
| | - Zhidong Yang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou, Guangzhou, 510405, China
| | - Xiaobing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine Guangzhou, Guangzhou, 510405, China. .,Laboratory Affiliated to National Key Discipline of Orthopaedic and Traumatology of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|