1
|
Vernia F, Ribichini E, Burrelli Scotti G, Latella G. Nutritional Deficiencies and Reduced Bone Mineralization in Ulcerative Colitis. J Clin Med 2025; 14:3202. [PMID: 40364233 PMCID: PMC12072929 DOI: 10.3390/jcm14093202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/03/2025] [Accepted: 05/04/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Inadequate dietary intake of vitamin D, vitamin K, and calcium, as well as sub-optimal sunlight exposure, can lead to bone loss in the general population, and more so in patients with ulcerative colitis, who are burdened by additional predisposing factors for osteoporosis, such as chronic inflammation and cortisone use. However, micronutrient deficiencies, if present, are easily corrected by nutritional intervention. While the relation between calcium and vitamin D and bone metabolism is well known, fewer data are available for vitamin K, for both healthy individuals and patients. The aim of this review is to provide an overview of recent reports focusing on nutritional deficits relevant to the development of osteoporosis/osteopenia in patients affected by ulcerative colitis. Methods: A systematic electronic search of the English literature up to January 2025 was performed using Medline and the Cochrane Library. Results: Despite being central in bone mineralization, data on dietary calcium intake in ulcerative colitis are relatively scarce, deriving mostly from mixed inflammatory bowel disease cohorts. Although lower than controls, dietary calcium intake approaches the recommended daily allowance, which establishes the necessary daily intake of nutrients. Conversely, vitamin D and vitamin K deficiencies are highly prevalent in ulcerative colitis patients. The widely shared opinion that milk and lactose-containing foods, as well as vegetables, worsen diarrhea is a prime determinant of inadequate vitamin D and vitamin K intake. Conclusions: Increased awareness of the importance of nutrition and the common occurrence of nutritional deficits represents the first step for the development of dietary intervention strategies to counteract the increased risk of osteoporosis in ulcerative colitis patients.
Collapse
Affiliation(s)
- Filippo Vernia
- Department of Life, Health, and Environmental Sciences, Division of Gastroenterology, Hepatology, and Nutrition, University of L’Aquila, Piazza S. Tommasi, 1-Coppito, 67100 L’Aquila, Italy;
| | - Emanuela Ribichini
- Department of Translational and Precision Medicine, Sapienza University, 00185 Rome, Italy; (E.R.); (G.B.S.)
| | - Giorgia Burrelli Scotti
- Department of Translational and Precision Medicine, Sapienza University, 00185 Rome, Italy; (E.R.); (G.B.S.)
| | - Giovanni Latella
- Department of Life, Health, and Environmental Sciences, Division of Gastroenterology, Hepatology, and Nutrition, University of L’Aquila, Piazza S. Tommasi, 1-Coppito, 67100 L’Aquila, Italy;
| |
Collapse
|
2
|
Li L, Lin R, Xu Y, Li L, Pan Z, Huang J. FoxA1 knockdown promotes BMSC osteogenesis in part by activating the ERK1/2 signaling pathway and preventing ovariectomy-induced bone loss. Sci Rep 2025; 15:4594. [PMID: 39920313 PMCID: PMC11806018 DOI: 10.1038/s41598-025-88658-1] [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: 06/02/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
The influence of deep learning in the medical and molecular biology sectors is swiftly growing and holds the potential to improve numerous crucial domains. Osteoporosis is a significant global health issue, and the current treatment options are highly restricted. Transplanting genetically engineered MSCs has been acknowledged as a highly promising therapy for osteoporosis. We utilized a random walk-based technique to discern genes associated with ossification. The osteogenic value of these genes was assessed on the basis of information found in published scientific literature. GO enrichment analysis of these genes was performed to determine if they were enriched in any certain function. Immunohistochemical and western blot techniques were used to identify and measure protein expression. The expression of genes involved in osteogenic differentiation was examined via qRT‒PCR. Lentiviral transfection was utilized to suppress the expression of the FOXA1 gene in hBMSCs. An in vivo mouse model of ovariectomy was created, and radiographic examination was conducted to confirm the impact of FOXA1 knockdown on osteoporosis. The osteogenic score of each gene was calculated by assessing its similarity to osteo-specific genes. The majority of the genes with the highest rankings were linked with osteogenic differentiation, indicating that our approach is useful for identifying genes associated with ossification. GO enrichment analysis revealed that these pathways are enriched primarily in bone-related processes. FOXA1 is a crucial transcription factor that controls the process of osteogenic differentiation, as indicated by similarity analysis. FOXA1 was significantly increased in those with osteoporosis. Downregulation of FOXA1 markedly augmented the expression of osteoblast-specific genes and proteins, activated the ERK1/2 signaling pathway, intensified ALP activity, and promoted mineral deposition. In addition, excessive expression of FOXA1 significantly reduced ALP activity and mineral deposits. Using a mouse model in which the ovaries were surgically removed, researchers reported that suppressing the FOXA1 gene in bone marrow stem cells (BMSCs) prevented the loss of bone density caused by ovariectomy. This finding was confirmed by analyzing the bone structure via micro-CT. Furthermore, our approach can distinguish genes that exhibit osteogenic differentiation characteristics. This ability can aid in the identification of novel genes associated with osteogenic differentiation, which can be utilized in the treatment of osteoporosis. Computational and laboratory evidence indicates that reducing the expression of FOXA1 enhances the process of bone formation in bone marrow-derived mesenchymal stem cells (BMSCs) and may serve as a promising approach to prevent osteoporosis.
Collapse
Affiliation(s)
- Lijun Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Zhejiang Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases, Hangzhou, 310016, Zhejiang, China
| | - Renjin Lin
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hang Zhou, China
| | - Yang Xu
- Thoracic Surgery Department of Zhejiang Cancer Hospital, Hangzhou, China
| | - Lingdi Li
- Department of Medical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Zhijun Pan
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China.
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hang Zhou, China.
| | - Jian Huang
- Department of Ultrasound, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
3
|
Gerini G, Traversa A, Cece F, Cassandri M, Pontecorvi P, Camero S, Nannini G, Romano E, Marampon F, Venneri MA, Ceccarelli S, Angeloni A, Amedei A, Marchese C, Megiorni F. Deciphering the Transcriptional Metabolic Profile of Adipose-Derived Stem Cells During Osteogenic Differentiation and Epigenetic Drug Treatment. Cells 2025; 14:135. [PMID: 39851564 PMCID: PMC11763738 DOI: 10.3390/cells14020135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 01/26/2025] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) are commonly employed in clinical treatment for various diseases due to their ability to differentiate into multi-lineage and anti-inflammatory/immunomodulatory properties. Preclinical studies support their use for bone regeneration, healing, and the improvement of functional outcomes. However, a deeper understanding of the molecular mechanisms underlying ASC biology is crucial to identifying key regulatory pathways that influence differentiation and enhance regenerative potential. In this study, we employed the NanoString nCounter technology, an advanced multiplexed digital counting method of RNA molecules, to comprehensively characterize differentially expressed transcripts involved in metabolic pathways at distinct time points in osteogenically differentiating ASCs treated with or without the pan-DNMT inhibitor RG108. In silico annotation and gene ontology analysis highlighted the activation of ethanol oxidation, ROS regulation, retinoic acid metabolism, and steroid hormone metabolism, as well as in the metabolism of lipids, amino acids, and nucleotides, and pinpointed potential new osteogenic drivers like AOX1 and ADH1A. RG108-treated cells, in addition to the upregulation of the osteogenesis-related markers RUNX2 and ALPL, showed statistically significant alterations in genes implicated in transcriptional control (MYCN, MYB, TP63, and IRF1), ethanol oxidation (ADH1C, ADH4, ADH6, and ADH7), and glucose metabolism (SLC2A3). These findings highlight the complex interplay of the metabolic, structural, and signaling pathways that orchestrate osteogenic differentiation. Furthermore, this study underscores the potential of epigenetic drugs like RG108 to enhance ASC properties, paving the way for more effective and personalized cell-based therapies for bone regeneration.
Collapse
Affiliation(s)
- Giulia Gerini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Alice Traversa
- Department of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy; (A.T.); (S.C.)
| | - Fabrizio Cece
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Matteo Cassandri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Paola Pontecorvi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Simona Camero
- Department of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy; (A.T.); (S.C.)
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy; (G.N.); (A.A.)
| | - Enrico Romano
- Department of Sense Organs, Sapienza University of Rome, 00161 Rome, Italy;
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00161 Rome, Italy;
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Antonio Angeloni
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy; (G.N.); (A.A.)
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| |
Collapse
|
4
|
Sumague TS, Niazy AA, Lambarte RNA, Nafisah IA, Gusnanto A. Influence of budesonide and fluticasone propionate in the anti-osteoporotic potential in human bone marrow-derived mesenchymal stem cells via stimulation of osteogenic differentiation. Heliyon 2024; 10:e39475. [PMID: 39497989 PMCID: PMC11532851 DOI: 10.1016/j.heliyon.2024.e39475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 09/24/2024] [Accepted: 10/15/2024] [Indexed: 11/07/2024] Open
Abstract
Osteoporosis is a prevalent bone condition with adverse effects observed in patients undergoing long-term glucocorticoid therapy, resulting in bone demineralization and tissue loss. There has been limited studies on the global response to dexamethasone in terms of comparing its expression profile to other common glucocorticoids during osteogenic differentiation. This study focused on the downregulated gene expression profile of glucocorticoid compounds; dexamethasone, budesonide, and fluticasone propionate, during osteogenic differentiation to elucidate the related target genes and pathways associated with the anti-osteoporotic potential of telomerase-immortalized human bone marrow-derived mesenchymal stem cells using a bioinformatics approach. Based on gene expression microarrays experiments and bioinformatics analysis, several key genes involved in the regulation of osteogenic differentiation and osteoporosis development in mesenchymal stem cells that were targeted by these specific glucocorticoids were determined. Network analysis using GeneCards, OMIM, and CTD databases were performed and osteoporosis-related genes were identified. LIMMA and moderated Welch test R packages were performed to determine significant downregulated differentially expressed genes for each glucocorticoid treatment. A total of 479 (dexamethasone), 84 (budesonide), and 889 (fluticasone propionate) differentially expressed genes were identified for each glucocorticoid, of which 35 common genes overlapped. Enrichment pathway analysis was conducted using Metascape, and protein-protein interaction networks were constructed using the STRING database and Cytoscape software to determine potential target genes involved with osteoporosis. Enrichment pathway analysis revealed genes involved in 3 Reactome pathways namely cytokine signaling in immune system, immune system and the interferon alpha/beta signaling pathways and identified 10 hub genes based on the PPI network to determine potential target pathways associated with osteoporosis. These findings provide preliminary insights into the relationship between the key target genes of dexamethasone, budesonide, and fluticasone propionate, and the pathways associated with regulated osteoporosis metabolism during osteogenic differentiation.
Collapse
Affiliation(s)
- Terrence Suministrado Sumague
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, King Saud University Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Abdurahman A. Niazy
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, King Saud University Medical City, Riyadh, Kingdom of Saudi Arabia
- Department of Oral Medicine and Diagnostic Sciences, College of Dentistry, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Rhodanne Nicole A. Lambarte
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, King Saud University Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Ibrahim A. Nafisah
- Department of Statistics and Operations Research, College of Science, King Saud University, Riyadh, Saudi Arabia
| | | |
Collapse
|
5
|
Luo W, Zhang N, Wang Z, Chen H, Sun J, Yao C, Zhang Y. LncRNA USP2-AS1 facilitates the osteogenic differentiation of bone marrow mesenchymal stem cells by targeting KDM3A/ETS1/USP2 to activate the Wnt/β-catenin signaling pathway. RNA Biol 2024; 21:1-13. [PMID: 38131611 PMCID: PMC10761055 DOI: 10.1080/15476286.2023.2290771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 12/23/2023] Open
Abstract
Human bone marrow mesenchymal stem cells (HBMSCs) can promote new bone formation. Previous studies have proven the ability of long non-coding RNAs (lncRNAs) to modulate the osteogenic differentiation of mesenchymal stem cells. However, the molecular mechanism modulated by lncRNAs in affecting the osteogenic differentiation of HBMSCs remains largely unknown. Thus, this study aims to reveal the role of lncRNA ubiquitin-specific peptidase 2 antisense RNA 1 (USP2-AS1) in regulating the osteogenic differentiation of HBMSCs and investigate its regulatory mechanism. Through bioinformatics analysis and RT-qPCR, we confirmed that USP2-AS1 expression was increased in HBMSCs after culturing in osteogenic differentiation medium (OM-HBMSCs). Moreover, we uncovered that knockdown of USP2-AS1 inhibited the osteogenic differentiation of HBMSCs. Further exploration indicated that USP2-AS1 positively regulated the expression of its nearby gene USP2. Mechanistically, USP2-AS1 recruited lysine demethylase 3A (KDM3A) to stabilize ETS proto-oncogene 1 (ETS1), transcription factor that transcriptionally activated USP2. Additionally, USP2-induced Wnt/β-catenin signalling pathway activation via deubiquitination of β-catenin protein. In summary, our study proved that lncRNA USP2-AS1 facilitates the osteogenic differentiation of HBMSCs by targeting KDM3A/ETS1/USP2 axis to activate the Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Wanxin Luo
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Na Zhang
- Department of Endocrinology, Jiangxi Provincial People’s Hospital, Nanchang, Jiangxi, China
- Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Ziping Wang
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Hao Chen
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jie Sun
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Chen Yao
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yafeng Zhang
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
6
|
Chen D, Liu S, Chu X, Reiter J, Gao H, McGuire P, Yu X, Xuei X, Liu Y, Wan J, Fang F, Liu Y, Wang Y. Osteogenic Differentiation Potential of Mesenchymal Stem Cells Using Single Cell Multiomic Analysis. Genes (Basel) 2023; 14:1871. [PMID: 37895219 PMCID: PMC10606235 DOI: 10.3390/genes14101871] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Mesenchymal stem cells (MSC) are multipotent stem cells that can differentiate into multiple cell types, including osteoblasts, chondrocytes, and adipocytes. Osteoblast differentiation is reduced during osteoporosis development, resulting in reduced bone formation. Further, MSC isolated from different donors possess distinct osteogenic capacity. In this study, we used single-cell multiomic analysis to profile the transcriptome and epigenome of MSC from four healthy donors. Data were obtained from ~1300 to 1600 cells for each donor. These cells were clustered into four groups, indicating that MSC from different donors have distinct chromatin accessible regulatory elements for regulating gene expression. To investigate the mechanism by which MSC undergo osteogenic differentiation, we used the chromatin accessibility data from the single-cell multiome data to identify individual-specific enhancer-promoter pairs and evaluated the expression levels and activities of the transcriptional regulators. The MSC from four donors showed distinct differentiation potential into osteoblasts. MSC of donor 1 showed the largest average motif activities, indicating that MSC from donor 1 was most likely to differentiate into osteoblasts. The results of our validation experiments were consistent with the bioinformatics prediction. We also tested the enrichment of genome-wide association study (GWAS) signals of several musculoskeletal disease traits in the patient-specific chromatin accessible regions identified in the single-cell multiome data, including osteoporosis, osteopenia, and osteoarthritis. We found that osteoarthritis-associated variants were only enriched in the regions identified from donor 4. In contrast, osteoporosis and osteopenia variants were enriched in regions from donor 1 and least enriched in donor 4. Since osteoporosis and osteopenia are related to the density of bone cells, the enrichment of variants from these traits should be correlated with the osteogenic potential of MSC. In summary, this study provides large-scale data to link regulatory elements with their target genes to study the regulatory relationships during the differentiation of mesenchymal stem cells and provide a deeper insight into the gene regulatory mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Yue Wang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
7
|
Stoilov B, Truong VK, Gronthos S, Vasilev K. Noninvasive and Microinvasive Nanoscale Drug Delivery Platforms for Hard Tissue Engineering. ACS APPLIED BIO MATERIALS 2023; 6:2925-2943. [PMID: 37565698 DOI: 10.1021/acsabm.3c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Bone tissue plays a crucial role in protecting internal organs and providing structural support and locomotion of the body. Treatment of hard tissue defects and medical conditions due to physical injuries, genetic disorders, aging, metabolic syndromes, and infections is more often a complex and drawn out process. Presently, dealing with hard-tissue-based clinical problems is still mostly conducted via surgical interventions. However, advances in nanotechnology over the last decades have led to shifting trends in clinical practice toward noninvasive and microinvasive methods. In this review article, recent advances in the development of nanoscale platforms for bone tissue engineering have been reviewed and critically discussed to provide a comprehensive understanding of the advantages and disadvantages of noninvasive and microinvasive methods for treating medical conditions related to hard tissue regeneration and repair.
Collapse
Affiliation(s)
- Borislav Stoilov
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia 5042, Australia
| | - Vi Khanh Truong
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia 5042, Australia
| | - Stan Gronthos
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide/SAHMRI, North Terrace, Adelaide, South Australia 5001, Australia
| | - Krasimir Vasilev
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia 5042, Australia
| |
Collapse
|
8
|
Tong X, Zhang Y, Zhao Y, Li Y, Li T, Zou H, Yuan Y, Bian J, Liu Z, Gu J. Vitamin D Alleviates Cadmium-Induced Inhibition of Chicken Bone Marrow Stromal Cells' Osteogenic Differentiation In Vitro. Animals (Basel) 2023; 13:2544. [PMID: 37570352 PMCID: PMC10417335 DOI: 10.3390/ani13152544] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/18/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Vitamin D is a lipid soluble vitamin that is mostly used to treat bone metabolism-related diseases. In this study, the effect of Cd toxicity in vitro on osteogenic differentiation derived from BMSCs and the alleviating effect of lα, 25-(OH)2D3 were investigated. Cell index in real time was monitored using a Real-time cell analyzer (RTCA) system. The activity of alkaline phosphatase (ALP), and the calcified nodules and the distribution of Runx2 protein were detected using ALP staining, alizarin red staining, and immunofluorescence, respectively. Furthermore, the mitochondrial membrane potential and the apoptotic rate of BMSCs, the mRNA levels of RUNX2 and type Ⅰ collagen alpha2 (COL1A2) genes, and the protein expression of Col1 and Runx2 were detected using flow cytometry, qRT-PCR and western blot, respectively. The proliferation of BMSCs and osteogenic differentiation were enhanced after treatment with different concentrations of lα, 25-(OH)2D3 compared with the control group. However, 5 μmol/L Cd inhibited the proliferation of BMSCs. In addition, 10 nmol/L lα,25-(OH)2D3 attenuated the toxicity and the apoptosis of BMSCs treated by Cd, and also promoted the osteogenic differentiation including the activity of ALP, and the protein expression of Col1 and Runx2. lα, 25-(OH)2D3 can alleviate cadmium-induced osteogenic toxicity in White Leghorn chickens in vitro.
Collapse
Affiliation(s)
- Xishuai Tong
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.T.); (H.Z.); (Y.Y.); (J.B.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Ying Zhang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China;
| | - Yutian Zhao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.T.); (H.Z.); (Y.Y.); (J.B.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Yawen Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.T.); (H.Z.); (Y.Y.); (J.B.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Tan Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.T.); (H.Z.); (Y.Y.); (J.B.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Hui Zou
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.T.); (H.Z.); (Y.Y.); (J.B.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Yan Yuan
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.T.); (H.Z.); (Y.Y.); (J.B.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Jianchun Bian
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.T.); (H.Z.); (Y.Y.); (J.B.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Zongping Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.T.); (H.Z.); (Y.Y.); (J.B.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| | - Jianhong Gu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (X.T.); (H.Z.); (Y.Y.); (J.B.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, China
| |
Collapse
|
9
|
Liu L, Rosen CJ. New Insights into Calorie Restriction Induced Bone Loss. Endocrinol Metab (Seoul) 2023; 38:203-213. [PMID: 37150516 PMCID: PMC10164494 DOI: 10.3803/enm.2023.1673] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/30/2023] [Indexed: 05/09/2023] Open
Abstract
Caloric restriction (CR) is now a popular lifestyle choice due to its ability in experimental animals to improve lifespan, reduce body weight, and lessen oxidative stress. However, more and more emerging evidence suggests this treatment requires careful consideration because of its detrimental effects on the skeletal system. Experimental and clinical studies show that CR can suppress bone growth and raise the risk of fracture, but the specific mechanisms are poorly understood. Reduced mechanical loading has long been thought to be the primary cause of weight loss-induced bone loss from calorie restriction. Despite fat loss in peripheral depots with calorie restriction, bone marrow adipose tissue (BMAT) increases, and this may play a significant role in this pathological process. Here, we update recent advances in our understanding of the effects of CR on the skeleton, the possible pathogenic role of BMAT in CR-induced bone loss, and some strategies to mitigate any potential side effects on the skeletal system.
Collapse
Affiliation(s)
- Linyi Liu
- MaineHealth Institute for Research, Scarborough, ME, USA
| | | |
Collapse
|
10
|
Puts R, Khaffaf A, Shaka M, Zhang H, Raum K. Focused Low-Intensity Pulsed Ultrasound (FLIPUS) Mitigates Apoptosis of MLO-Y4 Osteocyte-like Cells. Bioengineering (Basel) 2023; 10:bioengineering10030387. [PMID: 36978778 PMCID: PMC10045139 DOI: 10.3390/bioengineering10030387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Long cytoplasmic processes of osteocytes orchestrate bone activity by integration of biochemical and mechanical signals and regulate load-induced bone adaptation. Low-Intensity Pulsed Ultrasound (LIPUS) is a clinically used technique for fracture healing that delivers mechanical impulses to the damaged bone tissue in a non-invasive and non-ionizing manner. The mechanism of action of LIPUS is still controversially discussed in the scientific community. In this study, the effect of focused LIPUS (FLIPUS) on the survival of starved MLO-Y4 osteocytes was investigated in vitro. Osteocytes stimulated for 10 min with FLIPUS exhibited extended dendrites, which formed frequent connections to neighboring cells and spanned longer distances. The sonicated cells displayed thick actin bundles and experienced increase in expression of connexin 43 (Cx43) proteins, especially on their dendrites, and E11 glycoprotein, which is responsible for the elongation of cellular cytoplasmic processes. After stimulation, expression of cell growth and survival genes as well as genes related to cell-cell communication was augmented. In addition, cell viability was improved after the sonication, and a decrease in ATP release in the medium was observed. In summary, FLIPUS mitigated apoptosis of starved osteocytes, which is likely related to the formation of the extensive dendritic network that ensured cell survival.
Collapse
Affiliation(s)
- Regina Puts
- Center for Biomedicine, Charité-Universitätsmedizin, 12203 Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Charité-Universitätsmedizin, 13353 Berlin, Germany
| | - Aseel Khaffaf
- Center for Biomedicine, Charité-Universitätsmedizin, 12203 Berlin, Germany
| | - Maria Shaka
- Center for Biomedicine, Charité-Universitätsmedizin, 12203 Berlin, Germany
| | - Hui Zhang
- Center for Biomedicine, Charité-Universitätsmedizin, 12203 Berlin, Germany
| | - Kay Raum
- Center for Biomedicine, Charité-Universitätsmedizin, 12203 Berlin, Germany
| |
Collapse
|
11
|
Jin Q, Liu Y, Zhang Z, Wen X, Chen Z, Tian H, Kang Z, Wu X, Xu H. MYC promotes fibroblast osteogenesis by regulating ALP and BMP2 to participate in ectopic ossification of ankylosing spondylitis. Arthritis Res Ther 2023; 25:28. [PMID: 36803548 PMCID: PMC9942334 DOI: 10.1186/s13075-023-03011-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 02/09/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Ectopic ossification is an important cause of disability in patients with ankylosing spondylitis (AS). Whether fibroblasts can transdifferentiate into osteoblasts and contribute to ossification remains unknown. This study aims to investigate the role of stem cell transcription factors (POU5F1, SOX2, KLF4, MYC, etc.) of fibroblasts in ectopic ossification in patients with AS. METHODS Primary fibroblasts were isolated from the ligaments of patients with AS or osteoarthritis (OA). In an in vitro study, primary fibroblasts were cultured in osteogenic differentiation medium (ODM) to induce ossification. The level of mineralization was assessed by mineralization assay. The mRNA and protein levels of stem cell transcription factors were measured by real-time quantitative PCR (q-PCR) and western blotting. MYC was knocked down by infecting primary fibroblasts with lentivirus. The interactions between stem cell transcription factors and osteogenic genes were analysed by chromatin immunoprecipitation (ChIP). Recombinant human cytokines were added to the osteogenic model in vitro to evaluate their role in ossification. RESULTS We found that MYC was elevated significantly in the process of inducing primary fibroblasts to differentiate into osteoblasts. In addition, the level of MYC was remarkably higher in AS ligaments than in OA ligaments. When MYC was knocked down, the expression of the osteogenic genes alkaline phosphatase (ALP) and bone morphogenic protein 2 (BMP2) was decreased, and the level of mineralization was reduced significantly. In addition, the ALP and BMP2 were confirmed to be the direct target genes of MYC. Furthermore, interferon-γ (IFN-γ), which showed high expression in AS ligaments, was found to promote the expression of MYC in fibroblasts in the process of ossification in vitro. CONCLUSIONS This study demonstrates the role of MYC in ectopic ossification. MYC may act as the critical bridge that links inflammation with ossification in AS, thus providing new insights into the molecular mechanisms of ectopic ossification in AS.
Collapse
Affiliation(s)
- Qianmei Jin
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Yaoyang Liu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Zhiguo Zhang
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Xingzhu Wen
- Department of General Surgery, 72nd Group Army Hospital, Huzhou University, Huzhou, 313000, Zhejiang, China
| | - Ziqiang Chen
- Department of Orthopaedics, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Haijun Tian
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zijian Kang
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Xin Wu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
- School of Medicine, Tsinghua University, Beijing, 100084, China.
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
12
|
Vitamin D and Bone: A Story of Endocrine and Auto/Paracrine Action in Osteoblasts. Nutrients 2023; 15:nu15030480. [PMID: 36771187 PMCID: PMC9919888 DOI: 10.3390/nu15030480] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Despite its rigid structure, the bone is a dynamic organ, and is highly regulated by endocrine factors. One of the major bone regulatory hormones is vitamin D. Its renal metabolite 1α,25-OH2D3 has both direct and indirect effects on the maintenance of bone structure in health and disease. In this review, we describe the underlying processes that are directed by bone-forming cells, the osteoblasts. During the bone formation process, osteoblasts undergo different stages which play a central role in the signaling pathways that are activated via the vitamin D receptor. Vitamin D is involved in directing the osteoblasts towards proliferation or apoptosis, regulates their differentiation to bone matrix producing cells, and controls the subsequent mineralization of the bone matrix. The stage of differentiation/mineralization in osteoblasts is important for the vitamin D effect on gene transcription and the cellular response, and many genes are uniquely regulated either before or during mineralization. Moreover, osteoblasts contain the complete machinery to metabolize active 1α,25-OH2D3 to ensure a direct local effect. The enzyme 1α-hydroxylase (CYP27B1) that synthesizes the active 1α,25-OH2D3 metabolite is functional in osteoblasts, as well as the enzyme 24-hydroxylase (CYP24A1) that degrades 1α,25-OH2D3. This shows that in the past 100 years of vitamin D research, 1α,25-OH2D3 has evolved from an endocrine regulator into an autocrine/paracrine regulator of osteoblasts and bone formation.
Collapse
|
13
|
Hao RH, Guo Y, Wang C, Chen F, Di CX, Dong SS, Cao QL, Guo J, Rong Y, Yao S, Zhu DL, Chen YX, Chen H, Yang TL. Lineage-specific rearrangement of chromatin loops and epigenomic features during adipocytes and osteoblasts commitment. Cell Death Differ 2022; 29:2503-2518. [PMID: 35906483 PMCID: PMC9751090 DOI: 10.1038/s41418-022-01035-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 01/31/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) can be differentiated into adipocytes and osteoblasts. The processes are driven by the rewiring of chromatin architectures and transcriptomic/epigenomic changes. Here, we induced hMSCs to adipogenic and osteogenic differentiation, and performed 2 kb resolution Hi-C experiments for chromatin loops detection. We also generated matched RNA-seq, ChIP-seq and ATAC-seq data for integrative analysis. After comprehensively comparing adipogenesis and osteogenesis, we quantitatively identified lineage-specific loops and screened out lineage-specific enhancers and open chromatin. We reveal that lineage-specific loops can activate gene expression and facilitate cell commitment through combining enhancers and accessible chromatin in a lineage-specific manner. We finally proposed loop-mediated regulatory networks and identified the controlling factors for adipocytes and osteoblasts determination. Functional experiments validated the lineage-specific regulation networks towards IRS2 and RUNX2 that are associated with adipogenesis and osteogenesis, respectively. These results are expected to help better understand the chromatin conformation determinants of hMSCs fate commitment.
Collapse
Affiliation(s)
- Ruo-Han Hao
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Yan Guo
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Chen Wang
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Fei Chen
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Chen-Xi Di
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Shan-Shan Dong
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Qi-Long Cao
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- Research and Development Department, Qingdao Haier Biotech Co. Ltd, Qingdao, Shandong, 266109, P. R. China
| | - Jing Guo
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Yu Rong
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Shi Yao
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, P. R. China
| | - Dong-Li Zhu
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Yi-Xiao Chen
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, P. R. China
| | - Hao Chen
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China
| | - Tie-Lin Yang
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China.
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, P. R. China.
| |
Collapse
|
14
|
Wang J, Zhao J, Hu P, Gao L, Tian S, He Z. Long Non-coding RNA HOTAIR in Central Nervous System Disorders: New Insights in Pathogenesis, Diagnosis, and Therapeutic Potential. Front Mol Neurosci 2022; 15:949095. [PMID: 35813070 PMCID: PMC9259972 DOI: 10.3389/fnmol.2022.949095] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/07/2022] [Indexed: 01/17/2023] Open
Abstract
Central nervous system (CNS) disorders, such as ischemic stroke, neurodegenerative diseases, multiple sclerosis, traumatic brain injury, and corresponding neuropathological changes, often lead to death or long-term disability. Long non-coding RNA (lncRNA) is a class of non-coding RNA with a transcription length over 200 nt and transcriptional regulation. lncRNA is extensively involved in physiological and pathological processes through epigenetic, transcription, and post-transcriptional regulation. Further, dysregulated lncRNA is closely related to the occurrence and development of human diseases, including CNS disorders. HOX Transcript antisense RNA (HOTAIR) is the first discovered lncRNA with trans-transcriptional regulation. Recent studies have shown that HOTAIR may participate in the regulation of the occurrence and development of CNS disorders. In addition, HOTAIR has the potential to become a new biomarker for the diagnosis and prognosis assessment of CNS disorders and even provide a new therapeutic target for CNS disorders. Here, we reviewed the research results of HOTAIR in CNS disorders to provide new insights into the pathogenesis, diagnostic value, and therapeutic target potential of HOTAIR in human CNS disorders.
Collapse
Affiliation(s)
- Jialu Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jiuhan Zhao
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Pan Hu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Lianbo Gao
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shen Tian
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhenwei He
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Zhenwei He,
| |
Collapse
|
15
|
Liu J, Gan L, Ma B, He S, Wu P, Li H, Xiong J. Alterations in chromatin accessibility during osteoblast and adipocyte differentiation in human mesenchymal stem cells. BMC Med Genomics 2022; 15:17. [PMID: 35101056 PMCID: PMC8802426 DOI: 10.1186/s12920-022-01168-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/27/2022] [Indexed: 02/06/2023] Open
Abstract
Although differential expression of genes is apparent during the adipogenic/osteogenic differentiation of marrow mesenchymal stem cells (MSCs), it is not known whether this is associated with changes in chromosomal structure. In this study, we used ATAC-sequencing technology to observe variations in chromatin assembly during the early stages of MSC differentiation. This showed significant changes in the number and distribution of chromosome accessibility at different time points of adipogenic/osteogenic differentiation. Sequencing of differential peaks indicated alterations in transcription factor motifs involved in MSC differentiation. Gene Ontology (GO) and pathway analysis indicated that changes in biological function resulted from the alterations in chromatin accessibility. We then integrated ATAC-seq and RNA-seq and found that only a small proportion of the overlapping genes were screened out from ATAC-seq and RNA-seq overlapping. Through GO and pathway analysis of these overlapped genes, we not only observed some known biological functions related to adipogenic/osteogenic differentiation but also noticed some unusual biological clustering during MSC differentiation. In summary, our work not only presents the landscape of chromatin accessibility of MSC during differentiation but also helps to further our understanding of the underlying mechanisms of gene expression in these processes.
Collapse
Affiliation(s)
- Jianyun Liu
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Lijun Gan
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Baichen Ma
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Shan He
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Ping Wu
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Huiming Li
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Jianjun Xiong
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China.
| |
Collapse
|
16
|
Zhang Q, Dong J, Zhang P, Zhou D, Liu F. Dynamics of Transcription Factors in Three Early Phases of Osteogenic, Adipogenic, and Chondrogenic Differentiation Determining the Fate of Bone Marrow Mesenchymal Stem Cells in Rats. Front Cell Dev Biol 2021; 9:768316. [PMID: 34765608 PMCID: PMC8576568 DOI: 10.3389/fcell.2021.768316] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022] Open
Abstract
The imbalance of osteogenic, adipogenic, and chondrogenic differentiation in bone marrow mesenchymal stem cells (BMSCs) occurred in multiple age-related degenerative diseases such as osteoporosis and osteoarthritis. In order to improve our understanding and control of multi-directional differentiation of BMSCs in rats, using high-throughput sequencing, we identified key gene regulatory events in the early stages of lineage commitment. Data analysis revealed two transcription factors (TFs, Tsc22d3, and Epas1) with elevated expression throughout the initiation of differentiation (3 h), lineage acquisition (12 h), and early lineage progression (72 h) of three-directional differentiation. For osteogenic differentiation, 792, 1,042, and 638 differentially expressed genes including 48, 59, and 34 TFs were identified at three time points, respectively. Moreover, the functional analysis demonstrated that 4, 12, and 5 TFs were only differentially expressed during osteogenic differentiation at 3, 12, and 72 h, respectively, and not during other two-directional differentiation. Hopx showed enhanced expression throughout three early phases during the osteogenic differentiation but no significant change in other two-directional differentiation. A similar pattern of Gbx2 expression occurred in chondrogenic differentiation. Thus, Hopx and other early responder TFs may control the osteogenic cell fate of BMSCs and participate in the development of osteoporosis. Gbx2 and other early responder TFs should be considered in mechanistic models that clarify cartilage-anabolic changes in the clinical progression of osteoarthritis.
Collapse
Affiliation(s)
| | | | | | | | - Fanxiao Liu
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
17
|
Ziemińska M, Sieklucka B, Pawlak K. Vitamin K and D Supplementation and Bone Health in Chronic Kidney Disease-Apart or Together? Nutrients 2021; 13:809. [PMID: 33804453 PMCID: PMC7999920 DOI: 10.3390/nu13030809] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/15/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
Vitamin K (VK) and vitamin D (VD) deficiency/insufficiency is a common feature of chronic kidney disease (CKD), leading to impaired bone quality and a higher risk of fractures. CKD patients, with disturbances in VK and VD metabolism, do not have sufficient levels of these vitamins for maintaining normal bone formation and mineralization. So far, there has been no consensus on what serum VK and VD levels can be considered sufficient in this particular population. Moreover, there are no clear guidelines how supplementation of these vitamins should be carried out in the course of CKD. Based on the existing results of preclinical studies and clinical evidence, this review intends to discuss the effect of VK and VD on bone remodeling in CKD. Although the mechanisms of action and the effects of these vitamins on bone are distinct, we try to find evidence for synergy between them in relation to bone metabolism, to answer the question of whether combined supplementation of VK and VD will be more beneficial for bone health in the CKD population than administering each of these vitamins separately.
Collapse
Affiliation(s)
- Marta Ziemińska
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, 15-222 Bialystok, Poland;
| | - Beata Sieklucka
- Department of Pharmacodynamics, Medical University of Bialystok, 15-222 Bialystok, Poland;
| | - Krystyna Pawlak
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, 15-222 Bialystok, Poland;
| |
Collapse
|
18
|
Gaus S, Li H, Li S, Wang Q, Kottek T, Hahnel S, Liu X, Deng Y, Ziebolz D, Haak R, Schmalz G, Liu L, Savkovic V, Lethaus B. Shared Genetic and Epigenetic Mechanisms between the Osteogenic Differentiation of Dental Pulp Stem Cells and Bone Marrow Stem Cells. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6697810. [PMID: 33628811 PMCID: PMC7884974 DOI: 10.1155/2021/6697810] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/04/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To identify the shared genetic and epigenetic mechanisms between the osteogenic differentiation of dental pulp stem cells (DPSC) and bone marrow stem cells (BMSC). MATERIALS AND METHODS The profiling datasets of miRNA expression in the osteogenic differentiation of mesenchymal stem cells from the dental pulp (DPSC) and bone marrow (BMSC) were searched in the Gene Expression Omnibus (GEO) database. The differential expression analysis was performed to identify differentially expressed miRNAs (DEmiRNAs) dysregulated in DPSC and BMSC osteodifferentiation. The target genes of the DEmiRNAs that were dysregulated in DPSC and BMSC osteodifferentiation were identified, followed by the identification of the signaling pathways and biological processes (BPs) of these target genes. Accordingly, the DEmiRNA-transcription factor (TFs) network and the DEmiRNAs-small molecular drug network involved in the DPSC and BMSC osteodifferentiation were constructed. RESULTS 16 dysregulated DEmiRNAs were found to be overlapped in the DPSC and BMSC osteodifferentiation, including 8 DEmiRNAs with a common expression pattern (8 upregulated DEmiRNAs (miR-101-3p, miR-143-3p, miR-145-3p/5p, miR-19a-3p, miR-34c-5p, miR-3607-3p, miR-378e, miR-671-3p, and miR-671-5p) and 1 downregulated DEmiRNA (miR-671-3p/5p)), as well as 8 DEmiRNAs with a different expression pattern (i.e., miR-1273g-3p, miR-146a-5p, miR-146b-5p, miR-337-3p, miR-382-3p, miR-4508, miR-4516, and miR-6087). Several signaling pathways (TNF, mTOR, Hippo, neutrophin, and pathways regulating pluripotency of stem cells), transcription factors (RUNX1, FOXA1, HIF1A, and MYC), and small molecule drugs (curcumin, docosahexaenoic acid (DHA), vitamin D3, arsenic trioxide, 5-fluorouracil (5-FU), and naringin) were identified as common regulators of both the DPSC and BMSC osteodifferentiation. CONCLUSION Common genetic and epigenetic mechanisms are involved in the osteodifferentiation of DPSCs and BMSCs.
Collapse
Affiliation(s)
- Sebastian Gaus
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, Leipzig 04103, Germany
| | - Hanluo Li
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, Leipzig 04103, Germany
| | - Simin Li
- Department of Cariology, Endodontology and Periodontology, University Leipzig, Liebigstr. 12, Leipzig 04103, Germany
| | - Qian Wang
- Department of Central Laboratory, Taian Central Hospital, Longtan Road No. 29, Taian, 271000 Shandong Province, China
| | - Tina Kottek
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, Leipzig 04103, Germany
| | - Sebastian Hahnel
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, Leipzig 04103, Germany
| | - Xiangqiong Liu
- Department of Molecular Cell Biology, Beijing Tibetan Hospital, China Tibetology Research Center, 218 Anwaixiaoguanbeili Street, Chaoyang, Beijing 100029, China
| | - Yupei Deng
- Department of Molecular Cell Biology, Beijing Tibetan Hospital, China Tibetology Research Center, 218 Anwaixiaoguanbeili Street, Chaoyang, Beijing 100029, China
| | - Dirk Ziebolz
- Department of Cariology, Endodontology and Periodontology, University Leipzig, Liebigstr. 12, Leipzig 04103, Germany
| | - Rainer Haak
- Department of Cariology, Endodontology and Periodontology, University Leipzig, Liebigstr. 12, Leipzig 04103, Germany
| | - Gerhard Schmalz
- Department of Cariology, Endodontology and Periodontology, University Leipzig, Liebigstr. 12, Leipzig 04103, Germany
| | - Lei Liu
- Department of Neurology, Shandong Provincial Third Hospital, Cheeloo Chollege of Medicine, Shandong University, Jinan, 100191 Shandong Province, China
| | - Vuk Savkovic
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, Leipzig 04103, Germany
| | - Bernd Lethaus
- Department of Cranio Maxillofacial Surgery, University Clinic Leipzig, Liebigstr. 12, Leipzig 04103, Germany
| |
Collapse
|
19
|
Wu J, Cai P, Lu Z, Zhang Z, He X, Zhu B, Zheng L, Zhao J. Identification of potential specific biomarkers and key signaling pathways between osteogenic and adipogenic differentiation of hBMSCs for osteoporosis therapy. J Orthop Surg Res 2020; 15:437. [PMID: 32967719 PMCID: PMC7510089 DOI: 10.1186/s13018-020-01965-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022] Open
Abstract
Background The differentiation of bone mesenchymal stem cells (BMSCs) into adipogenesis (AD) rather than osteogenesis (OS) is an important pathological feature of osteoporosis. Illuminating the detailed mechanisms of the differentiation of BMSCs into OS and AD would contribute to the interpretation of osteoporosis pathology. Methods To identify the regulated mechanism in lineage commitment of the BMSCs into OS and AD in the early stages, the gene expression profiles with temporal series were downloaded to reveal the distinct fates when BMSCs adopt a committed lineage. For both OS and AD lineages, the profiles of days 2–4 were compared with day 0 to screen the differentially expressed genes (DEGs), respectively. Next, the functional enrichment analysis was utilized to find out the biological function, and protein-protein interaction network to predict the central genes. Finally, experiments were performed to verify our finding. Results FoxO signaling pathway with central genes like FoxO3, IL6, and CAT is the crucial mechanism of OS, while Rap1 signaling pathway of VEGFA and FGF2 enrichment is more significant for AD. Besides, PI3K-Akt signaling pathway might serve as the latent mechanism about the initiation of differentiation of BMSCs into multiple lineages. Conclusion Above hub genes and early-responder signaling pathways control osteogenic and adipogenic fates of BMSCs, which maybe mechanistic models clarifying the changes of bone metabolism in the clinical progress of osteoporosis. The findings provide a crucial reference for the prevention and therapy of osteoporosis.
Collapse
Affiliation(s)
- Jianjun Wu
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Peian Cai
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Zhenhui Lu
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Zhi Zhang
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Xixi He
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Bikang Zhu
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China. .,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China. .,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China. .,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China. .,Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
20
|
Jacques C, Tesfaye R, Lavaud M, Georges S, Baud’huin M, Lamoureux F, Ory B. Implication of the p53-Related miR-34c, -125b, and -203 in the Osteoblastic Differentiation and the Malignant Transformation of Bone Sarcomas. Cells 2020; 9:cells9040810. [PMID: 32230926 PMCID: PMC7226610 DOI: 10.3390/cells9040810] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023] Open
Abstract
The formation of the skeleton occurs throughout the lives of vertebrates and is achieved through the balanced activities of two kinds of specialized bone cells: the bone-forming osteoblasts and the bone-resorbing osteoclasts. Impairment in the remodeling processes dramatically hampers the proper healing of fractures and can also result in malignant bone diseases such as osteosarcoma. MicroRNAs (miRNAs) are a class of small non-coding single-strand RNAs implicated in the control of various cellular activities such as proliferation, differentiation, and apoptosis. Their post-transcriptional regulatory role confers on them inhibitory functions toward specific target mRNAs. As miRNAs are involved in the differentiation program of precursor cells, it is now well established that this class of molecules also influences bone formation by affecting osteoblastic differentiation and the fate of osteoblasts. In response to various cell signals, the tumor-suppressor protein p53 activates a huge range of genes, whose miRNAs promote genomic-integrity maintenance, cell-cycle arrest, cell senescence, and apoptosis. Here, we review the role of three p53-related miRNAs, miR-34c, -125b, and -203, in the bone-remodeling context and, in particular, in osteoblastic differentiation. The second aim of this study is to deal with the potential implication of these miRNAs in osteosarcoma development and progression.
Collapse
|
21
|
A Rationale for the Use of Clotted Vertebral Bone Marrow to Aid Tissue Regeneration Following Spinal Surgery. Sci Rep 2020; 10:4115. [PMID: 32139727 PMCID: PMC7058026 DOI: 10.1038/s41598-020-60934-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/19/2020] [Indexed: 12/25/2022] Open
Abstract
Vertebral body bone marrow aspirate (V-BMA), easily accessible simultaneously with the preparation of the site for pedicle screw insertion during spinal procedures, is becoming an increasingly used cell therapy approach in spinal surgery. However, the main drawbacks for V-BMA use are the lack of a standardized procedure and of a structural texture with the possibility of diffusion away from the implant site. The aim of this study was to evaluate, characterize and compare the biological characteristics of MSCs from clotted V-BMA and MSCs from whole and concentrate V-BMAs. MSCs from clotted V-BMA showed the highest cell viability and growth factors expression (TGF-β, VEGF-A, FGF2), the greatest colony forming unit (CFU) potency, cellular homogeneity, ability to differentiate towards the osteogenic (COL1AI, TNFRSF11B, BGLAP) and chondrogenic phenotype (SOX9) and the lowest ability to differentiate toward the adipogenic lineage (ADIPOQ) in comparison to all the other culture conditions. Additionally, results revealed that MSCs, differently isolated, expressed different level of HOX and TALE signatures and that PBX1 and MEIS3 were down-regulated in MSCs from clotted V-BMA in comparison to concentrated one. The study demonstrated for the first time that the cellular source inside the clotted V-BMA showed the best biological properties, representing an alternative and advanced cell therapy approach for patients undergoing spinal surgery.
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW To review the available literature regarding a possible relationship between vitamin D and bone marrow adipose tissue (BMAT), and to identify future avenues of research that warrant attention. RECENT FINDINGS Results from in vivo animal and human studies all support the hypothesis that vitamin D can suppress BMAT expansion. This is achieved by antagonizing adipogenesis in bone marrow stromal cells, through inhibition of PPARγ2 activity and stimulation of pro-osteogenic Wnt signalling. However, our understanding of the functions of BMAT is still evolving, and studies on the role of vitamin D in modulating BMAT function are lacking. In addition, many diseases and chronic conditions are associated with low vitamin D status and low bone mineral density (BMD), but BMAT expansion has not been studied in these patient populations. Vitamin D suppresses BMAT expansion, but its role in modulating BMAT function is poorly understood.
Collapse
Affiliation(s)
- Hanel Sadie-Van Gijsen
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University Tygerberg Campus, Francie van Zijl Drive, PO Box 241, Parow, Cape Town, 8000, South Africa.
| |
Collapse
|
23
|
Abstract
Understanding of vitamin D physiology is important because about half of the population is being diagnosed with deficiency and treated with supplements. Clinical guidelines were developed based on observational studies showing an association between low serum levels and increased cardiovascular risk. However, new randomized controlled trials have failed to confirm any cardiovascular benefit from supplementation in the general population. A major concern is that excess vitamin D is known to cause calcific vasculopathy and valvulopathy in animal models. For decades, administration of vitamin D has been used in rodents as a reliable experimental model of vascular calcification. Technically, vitamin D is a misnomer. It is not a true vitamin because it can be synthesized endogenously through ultraviolet exposure of the skin. It is a steroid hormone that comes in 3 forms that are sequential metabolites produced by hydroxylases. As a fat-soluble hormone, the vitamin D-hormone metabolites must have special mechanisms for delivery in the aqueous bloodstream. Importantly, endogenously synthesized forms are carried by a binding protein, whereas dietary forms are carried within lipoprotein particles. This may result in distinct biodistributions for sunlight-derived versus supplement-derived vitamin D hormones. Because the cardiovascular effects of vitamin D hormones are not straightforward, both toxic and beneficial effects may result from current recommendations.
Collapse
Affiliation(s)
- Linda L Demer
- From the Departments of Medicine (L.L.D., J.J.H., Y.T.) .,Physiology (L.L.D., Y.T.).,Bioengineering (L.L.D.)
| | - Jeffrey J Hsu
- From the Departments of Medicine (L.L.D., J.J.H., Y.T.)
| | - Yin Tintut
- From the Departments of Medicine (L.L.D., J.J.H., Y.T.).,Physiology (L.L.D., Y.T.).,Orthopaedic Surgery (Y.T.), University of California, Los Angeles
| |
Collapse
|
24
|
Nah H, Lee D, Heo M, Lee JS, Lee SJ, Heo DN, Seong J, Lim HN, Lee YH, Moon HJ, Hwang YS, Kwon IK. Vitamin D-conjugated gold nanoparticles as functional carriers to enhancing osteogenic differentiation. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2019; 20:826-836. [PMID: 31489055 PMCID: PMC6713151 DOI: 10.1080/14686996.2019.1644193] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 05/09/2023]
Abstract
In an aging society, bone disorders such as osteopenia, osteoporosis, and degenerative arthritis cause serious public health problems. In order to solve these problems, researchers continue to develop therapeutic agents, increase the efficacy of developed therapeutic agents, and reduce side effects. Gold nanoparticles (GNPs) are widely used in tissue engineering applications as biosensors, drug delivery carriers, and bioactive materials. Their special surface property enables easy conjugation with ligands including functional groups such as thiols, phosphines, and amines. This creates an attractive advantage to GNPs for use in the bone tissue engineering field. However, GNPs alone are limited in their biological effects. In this study, we used thiol-PEG-vitamin D (SPVD) to conjugate vitamin D, an essential nutrient critical for maintaining normal skeletal homeostasis, to GNPs. To characterize vitamin D-conjugated GNPs (VGNPs), field emission transmission electron microscopy, energy dispersive X-ray spectroscopy, dynamic light scattering, and ultraviolet/visible absorption analysis were carried out. The developed VGNPs were well bound through the thiol groups between GNPs and vitamin D, and were fabricated in size of 60 nm. Moreover, to demonstrate VGNPs osteogenic differentiation effect, various assays were carried out through cell viability test, alkaline phosphatase assay, calcium deposition assay, real-time polymerase chain reaction, and immunofluorescence staining. As a result, the fabricated VGNPs were found to effectively enhance osteogenic differentiation of human adipose-derived stem cells (hADSCs) in vitro. Based on these results, VGNPs can be utilized as functional nanomaterials for bone regeneration in the tissue engineering field.
Collapse
Affiliation(s)
- Haram Nah
- Department of Dentistry, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Donghyun Lee
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Min Heo
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Jae Seo Lee
- Department of Dentistry, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Sang Jin Lee
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Dong Nyoung Heo
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Jeongmin Seong
- Department of Dental Hygiene, College of Health Science, Kangwon National University, Samcheok-si, Republic of Korea
| | - Ho-Nam Lim
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Yeon-Hee Lee
- Department of Orofacial Pain and Oral Medicine, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Ho-Jin Moon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Yu-Shik Hwang
- Department of Maxillofacial Biomedical Engineering, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
25
|
Zhang Z, Zhang X, Zhao D, Liu B, Wang B, Yu W, Li J, Yu X, Cao F, Zheng G, Zhang Y, Liu Y. TGF‑β1 promotes the osteoinduction of human osteoblasts via the PI3K/AKT/mTOR/S6K1 signalling pathway. Mol Med Rep 2019; 19:3505-3518. [PMID: 30896852 PMCID: PMC6471541 DOI: 10.3892/mmr.2019.10051] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 03/06/2019] [Indexed: 12/22/2022] Open
Abstract
Transforming growth factor β1 (TGF-β1) has been suggested to be a candidate cytokine in the field of bone tissue engineering. Cytokines serve important roles in tissue engineering, particularly in the repair of bone damage; however, the underlying molecular mechanisms remain unclear. In the present study, the effects of TGF-β1 on the osteogenesis and motility of hFOB1.19 human osteoblasts were demonstrated via the phenotype and gene expression of cells. Additionally, the role of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin/S6 kinase 1 (PI3K/AKT/mTOR/S6K1) signalling pathway in the effects of TGF-β1 on osteoblasts was investigated. It was demonstrated using Cell Counting Kit-8 and flow cytometry assays that the proliferation of human osteoblasts was promoted by 1 ng/ml TGF-β1. In addition, alkaline phosphatase activity, Alizarin red staining, scratch-wound and Transwell assays were conducted. It was revealed that osteogenesis and the migration of cells were regulated by TGF-β1 via the upregulation of osteogenic and migration-associated genes. Alterations in the expression of osteogenesis- and migration-associated genes were evaluated following pre-treatment with a PI3K/AKT inhibitor (LY294002) and an mTOR/S6K1 inhibitor (rapamycin), with or without TGF-β1. The results indicated that TGF-β1 affected the osteogenesis and mineralisation of osteoblasts via the PI3K/AKT signalling pathway. Furthermore, TGF-β1 exhibited effects on mTOR/S6K1 downstream of PI3K/AKT. The present study demonstrated that TGF-β1 promoted the proliferation, differentiation and migration of human hFOB1.19 osteoblasts, and revealed that TGF-β1 affected the biological activity of osteoblasts via the PI3K/AKT/mTOR/S6K1 signalling pathway. Our findings may provide novel insight to aid the development of bone tissue engineering methods for the treatment of bone injury.
Collapse
Affiliation(s)
- Zhaodong Zhang
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Xiuzhi Zhang
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Dewei Zhao
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Baoyi Liu
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Benjie Wang
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Weiting Yu
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Junlei Li
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Xiaobing Yu
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Fang Cao
- Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, P.R. China
| | - Guoshuang Zheng
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Yao Zhang
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Yupeng Liu
- Department of Orthopaedics, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| |
Collapse
|
26
|
Luttrell LM, Dar MS, Gesty-Palmer D, El-Shewy HM, Robinson KM, Haycraft CJ, Barth JL. Transcriptomic characterization of signaling pathways associated with osteoblastic differentiation of MC-3T3E1 cells. PLoS One 2019; 14:e0204197. [PMID: 30608923 PMCID: PMC6319725 DOI: 10.1371/journal.pone.0204197] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/16/2018] [Indexed: 12/21/2022] Open
Abstract
Bone remodeling involves the coordinated actions of osteoclasts, which resorb the calcified bony matrix, and osteoblasts, which refill erosion pits created by osteoclasts to restore skeletal integrity and adapt to changes in mechanical load. Osteoblasts are derived from pluripotent mesenchymal stem cell precursors, which undergo differentiation under the influence of a host of local and environmental cues. To characterize the autocrine/paracrine signaling networks associated with osteoblast maturation and function, we performed gene network analysis using complementary “agnostic” DNA microarray and “targeted” NanoString nCounter datasets derived from murine MC3T3-E1 cells induced to undergo synchronized osteoblastic differentiation in vitro. Pairwise datasets representing changes in gene expression associated with growth arrest (day 2 to 5 in culture), differentiation (day 5 to 10 in culture), and osteoblast maturation (day 10 to 28 in culture) were analyzed using Ingenuity Systems Pathways Analysis to generate predictions about signaling pathway activity based on the temporal sequence of changes in target gene expression. Our data indicate that some pathways involved in osteoblast differentiation, e.g. Wnt/β-catenin signaling, are most active early in the process, while others, e.g. TGFβ/BMP, cytokine/JAK-STAT and TNFα/RANKL signaling, increase in activity as differentiation progresses. Collectively, these pathways contribute to the sequential expression of genes involved in the synthesis and mineralization of extracellular matrix. These results provide insight into the temporal coordination and complex interplay between signaling networks controlling gene expression during osteoblast differentiation. A more complete understanding of these processes may aid the discovery of novel methods to promote osteoblast development for the treatment of conditions characterized by low bone mineral density.
Collapse
Affiliation(s)
- Louis M. Luttrell
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States of America
- * E-mail:
| | - Moahad S. Dar
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Diane Gesty-Palmer
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Hesham M. El-Shewy
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Katherine M. Robinson
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Courtney J. Haycraft
- Department of Biology, Mississippi College, Clinton, Mississippi, United States of America
| | - Jeremy L. Barth
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| |
Collapse
|
27
|
Abstract
Bone morphogenetic proteins (BMPs) constitute the largest subdivision of the transforming growth factor-β family of ligands. BMPs exhibit widespread utility and pleiotropic, context-dependent effects, and the strength and duration of BMP pathway signaling is tightly regulated at numerous levels via mechanisms operating both inside and outside the cell. Defects in the BMP pathway or its regulation underlie multiple human diseases of different organ systems. Yet much remains to be discovered about the BMP pathway in its original context, i.e., the skeleton. In this review, we provide a comprehensive overview of the intricacies of the BMP pathway and its inhibitors in bone development, homeostasis, and disease. We frame the content of the review around major unanswered questions for which incomplete evidence is available. First, we consider the gene regulatory network downstream of BMP signaling in osteoblastogenesis. Next, we examine why some BMP ligands are more osteogenic than others and what factors limit BMP signaling during osteoblastogenesis. Then we consider whether specific BMP pathway components are required for normal skeletal development, and if the pathway exerts endogenous effects in the aging skeleton. Finally, we propose two major areas of need of future study by the field: greater resolution of the gene regulatory network downstream of BMP signaling in the skeleton, and an expanded repertoire of reagents to reliably and specifically inhibit individual BMP pathway components.
Collapse
Affiliation(s)
- Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine , Indianapolis, Indiana ; and Department of Developmental Biology, Harvard School of Dental Medicine , Boston, Massachusetts
| | - Vicki Rosen
- Division of Biomedical Science, Marian University College of Osteopathic Medicine , Indianapolis, Indiana ; and Department of Developmental Biology, Harvard School of Dental Medicine , Boston, Massachusetts
| |
Collapse
|
28
|
Lu M, Zhao XH. The Growth Proliferation, Apoptotic Prevention, and Differentiation Induction of the Gelatin Hydrolysates from Three Sources to Human Fetal Osteoblasts (hFOB 1.19 Cells). Molecules 2018; 23:molecules23061287. [PMID: 29843361 PMCID: PMC6100253 DOI: 10.3390/molecules23061287] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 04/26/2018] [Accepted: 05/08/2018] [Indexed: 01/07/2023] Open
Abstract
Gelatins from the skin of bovine, porcine, and tilapia were hydrolyzed to three degrees of hydrolysis (DH) by alcalase, neutrase, and papain, respectively. These hydrolysates at 0.02⁻0.1 g/L promoted the growth of human fetal osteoblasts by 101.4⁻135.7%, while higher DH or using papain and tilapia gelatins resulted in higher proliferation. The hydrolysates from porcine and tilapia gelatins at 0.05 g/L prevented induced apoptosis (decreasing total apoptotic proportions from 28.4% or 35.2% to 10.3⁻17.5% or 16.0⁻23.6%), and had differentiation induction (increasing alkaline phosphatase activity by 126.9⁻246.7% in early differentiation stage, or enhancing osteocalcin production by 4.1⁻22.5% in later differentiation stage). These hydrolysates had a similar amino acid profile; however, tilapia gelatin hydrolysates by papain with DH 15.4% mostly displayed higher activity than others. Tilapia gelatin hydrolysate could up-regulate β-catenin, Wnt 3a, Wnt 10b, cyclin D1, and c-Myc expression at mRNA levels by 1.11⁻3.60 folds, but down-regulate GSK 3β expression by 0.98 fold. Of note, β-catenin in total cellular and nuclear protein was up-regulated by 1.14⁻1.16 folds but unchanged in cytoplasmic protein, Wnt 10b, cyclin D1, and c-Myc expression were up-regulated by 1.27⁻1.95 folds, whilst GSK 3β expression was down-regulated by 0.87 fold. Activation of Wnt/β-catenin pathway is suggested to mediate cell proliferation and differentiation.
Collapse
Affiliation(s)
- Ming Lu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | - Xin-Huai Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
- Department of Food Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
29
|
Kapil U, Pandey RM, Sharma B, Ramakrishnan L, Sharma N, Singh G, Sareen N. Prevalence of Vitamin D Deficiency in Children (6-18 years) Residing in Kullu and Kangra Districts of Himachal Pradesh, India. Indian J Pediatr 2018; 85:344-350. [PMID: 29292488 DOI: 10.1007/s12098-017-2577-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 12/14/2017] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To assess the prevalence of Vitamin D deficiency (VDD) and associated risk factors amongst children in the age group of 6-18 y residing at an altitude of 1000 mts and above. METHODS A community based cross-sectional study was conducted in the year 2015-2016. Two districts (namely: Kangra and Kullu) of Himachal Pradesh state, India was selected for the present study. In each district thirty clusters/schools were identified using Population Proportionate to Size (PPS) sampling methodology. In the identified school, all the children in schools were enlisted. Twenty children per school were selected by using random number tables. A total of 1222 children (Kangra: 610; Kullu: 612) in the age group of 6-18 y were enrolled. The data on socio economic status, physical activity and sunlight exposure was collected. The blood samples were collected and serum 25-hydroxyvitamin D, intact parathyroid hormone, serum calcium, phosphorous, albumin and alkaline phosphate were assessed using standard procedures. RESULTS Eighty one percent (Kangra) and 80.0% (Kullu) of school age children were found Vitamin D deficient as per serum 25(OH) D levels (less than 20 ng/ml). CONCLUSIONS A high prevalence of VDD was found in children residing in 2 districts located at high altitude regions of Himachal Pradesh, India.
Collapse
Affiliation(s)
- Umesh Kapil
- Department of Human Nutrition, All India Institute of Medical Sciences, New Delhi, India.
| | - Ravindra Mohan Pandey
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, India
| | - Brij Sharma
- Department of Gastroenterology, Indira Gandhi Medical College, Shimla, Himachal Pradesh, India
| | - Lakshmy Ramakrishnan
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Neetu Sharma
- Department of Physiology, Indira Gandhi Medical College, Shimla, Himachal Pradesh, India
| | - Gajendra Singh
- Department of Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| | - Neha Sareen
- Department of Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
30
|
Geraniin promotes osteoblast proliferation and differentiation via the activation of Wnt/β-catenin pathway. Biomed Pharmacother 2018; 99:319-324. [DOI: 10.1016/j.biopha.2018.01.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 12/30/2017] [Accepted: 01/03/2018] [Indexed: 12/31/2022] Open
|
31
|
Lopez-Baez JC, Simpson DJ, LLeras Forero L, Zeng Z, Brunsdon H, Salzano A, Brombin A, Wyatt C, Rybski W, Huitema LFA, Dale RM, Kawakami K, Englert C, Chandra T, Schulte-Merker S, Hastie ND, Patton EE. Wilms Tumor 1b defines a wound-specific sheath cell subpopulation associated with notochord repair. eLife 2018; 7:30657. [PMID: 29405914 PMCID: PMC5811212 DOI: 10.7554/elife.30657] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 02/02/2018] [Indexed: 12/18/2022] Open
Abstract
Regenerative therapy for degenerative spine disorders requires the identification of cells that can slow down and possibly reverse degenerative processes. Here, we identify an unanticipated wound-specific notochord sheath cell subpopulation that expresses Wilms Tumor (WT) 1b following injury in zebrafish. We show that localized damage leads to Wt1b expression in sheath cells, and that wt1b+cells migrate into the wound to form a stopper-like structure, likely to maintain structural integrity. Wt1b+sheath cells are distinct in expressing cartilage and vacuolar genes, and in repressing a Wt1b-p53 transcriptional programme. At the wound, wt1b+and entpd5+ cells constitute separate, tightly-associated subpopulations. Surprisingly, wt1b expression at the site of injury is maintained even into adult stages in developing vertebrae, which form in an untypical manner via a cartilage intermediate. Given that notochord cells are retained in adult intervertebral discs, the identification of novel subpopulations may have important implications for regenerative spine disorder treatments.
Collapse
Affiliation(s)
- Juan Carlos Lopez-Baez
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom.,CRUK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Daniel J Simpson
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Laura LLeras Forero
- Hubrecht Institute - KNAW & UMC Utrecht, Utrecht, Netherlands.,Faculty of Medicine, Institute for Cardiovascular Organogenesis and Regeneration, WWU Münster, Münster, Germany.,CiM Cluster of Excellence, Münster, Germany
| | - Zhiqiang Zeng
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom.,CRUK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Hannah Brunsdon
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom.,CRUK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Angela Salzano
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Alessandro Brombin
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom.,CRUK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Cameron Wyatt
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Witold Rybski
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom.,CRUK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Rodney M Dale
- Department of Biology, Loyola University Chicago, Chicago, United States
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan
| | - Christoph Englert
- Department of Molecular Genetics, Leibniz Institute for Age Research-Fritz Lipmann Institute, Jena, Germany.,Institute of Biochemistry and Biophysics, Friedrich-Schiller-University, Jena, Germany
| | - Tamir Chandra
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Stefan Schulte-Merker
- Hubrecht Institute - KNAW & UMC Utrecht, Utrecht, Netherlands.,Faculty of Medicine, Institute for Cardiovascular Organogenesis and Regeneration, WWU Münster, Münster, Germany.,CiM Cluster of Excellence, Münster, Germany
| | - Nicholas D Hastie
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - E Elizabeth Patton
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom.,CRUK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
32
|
Pahlevan Kakhki M, Nikravesh A, Shirvani Farsani Z, Sahraian MA, Behmanesh M. HOTAIR but not ANRIL long non-coding RNA contributes to the pathogenesis of multiple sclerosis. Immunology 2017; 153:479-487. [PMID: 29030863 DOI: 10.1111/imm.12850] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/26/2017] [Accepted: 10/06/2017] [Indexed: 01/17/2023] Open
Abstract
Studies have revealed that dysregulation in gene expression is one of the main aspects of multiple sclerosis (MS) pathogenesis. Although the molecular pathways underlying the immunomodulatory role of vitamin D (VD) in MS is not completely elucidated, VD has more recently become a topic of interest in immune regulation and is widely administered to patients with MS as an immunomodulatory supplement. Long non-coding RNAs (lncRNAs) are known to play important roles in regulation of gene expression via different mechanisms. Given that VD-related genes are regulated by epigenetic mechanisms, here we aimed to evaluate the role of VD in combination with HOTAIR and ANRIL lncRNAs using in vivo, in vitro and in silico experiments in MS pathogenesis. Our data revealed that HOTAIR but not ANRIL lncRNA is probably involved in the pathogenesis of MS and experimental autoimmune encephalomyelitis through an unclear mechanism and it seems that by affecting the expression, inflammation and VD can influence HOTAIR-related mechanisms, which require further study.
Collapse
Affiliation(s)
- Majid Pahlevan Kakhki
- Faculty of Biological Sciences, Department of Genetics, Tarbiat Modares University, Tehran, Iran
| | - Abbas Nikravesh
- Faculty of Medicine, Department of Medical Biotechnology & Molecular Sciences, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Zeinab Shirvani Farsani
- Faculty of Biological Sciences and Technology, Department of Cellular and Molecular Biology, Shahid Beheshti G.C., Tehran, Iran
| | - Mohammad Ali Sahraian
- MS Research Centre, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Behmanesh
- Faculty of Biological Sciences, Department of Genetics, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
33
|
Paracrine interactions between mesenchymal stem cells and macrophages are regulated by 1,25-dihydroxyvitamin D3. Sci Rep 2017; 7:14618. [PMID: 29097745 PMCID: PMC5668416 DOI: 10.1038/s41598-017-15217-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/23/2017] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSC) modulate the macrophage-mediated inflammatory response through the secretion of soluble factors. In addition to its classical effects on calcium homeostasis, 1,25-dihydroxyvitamin D3 (1,25D3) has emerged as an important regulator of the immune system. The present study investigates whether 1,25D3 modulates the paracrine interactions between MSC and macrophages. 1,25D3 stimulated MSC to produce PGE2 and VEGF and regulated the interplay between macrophages and MSC toward reduced pro-inflammatory cytokine production. Conditioned media (CM) from co-cultures of macrophages and MSC impaired MSC osteogenesis. However, MSC cultured in CM from 1,25D3-treated co-cultures showed increased matrix maturation and mineralization. Co-culturing MSC with macrophages prevented the 1,25D3-induced increase in RANKL levels, which correlated with up-regulation of OPG secretion. MSC seeding in three-dimensional (3D) substrates potentiated their immunomodulatory effects on macrophages. Exposure of 3D co-cultures to 1,25D3 further reduced the levels of soluble factors related to inflammation and chemotaxis. As a consequence of 1,25D3 treatment, the recruitment of monocytes toward CM of 3D co-cultures decreased, while the osteogenic maturation of MSC increased. These data add new insights into the pleiotropic effects of 1,25D3 on the crosstalk between MSC and macrophages and highlight the role of the hormone in bone regeneration.
Collapse
|
34
|
van Driel M, van Leeuwen JPTM. Vitamin D endocrinology of bone mineralization. Mol Cell Endocrinol 2017; 453:46-51. [PMID: 28606868 DOI: 10.1016/j.mce.2017.06.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 12/19/2022]
Abstract
Bone is a dynamic tissue that is strongly influenced by endocrine factors to restore the balance between bone resorption and bone formation. Bone formation involves the mineralization of the extracellular matrix formed by osteoblasts. In this process the role of vitamin D (1α,25(OH)2D3) is both direct and indirect. The direct effects are enabled via the Vitamin D Receptor (VDR); the outcome is dependent on the presence of other factors as well as origin of the osteoblasts, treatment procedures and species differences. Vitamin D stimulates mineralization of human osteoblasts but is often found inhibitory for mineralization of murine osteoblasts. In this review we will overview the current knowledge of the role of the vitamin D endocrine system in controlling the mineralization process in bone.
Collapse
Affiliation(s)
- Marjolein van Driel
- Department of Internal Medicine, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands.
| | | |
Collapse
|
35
|
Belair DG, Wolf CJ, Wood C, Ren H, Grindstaff R, Padgett W, Swank A, MacMillan D, Fisher A, Winnik W, Abbott BD. Engineering human cell spheroids to model embryonic tissue fusion in vitro. PLoS One 2017; 12:e0184155. [PMID: 28898253 PMCID: PMC5595299 DOI: 10.1371/journal.pone.0184155] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/19/2017] [Indexed: 01/06/2023] Open
Abstract
Epithelial-mesenchymal interactions drive embryonic fusion events during development, and perturbations of these interactions can result in birth defects. Cleft palate and neural tube defects can result from genetic defects or environmental exposures during development, yet very little is known about the effect of chemical exposures on fusion events during human development because of a lack of relevant and robust human in vitro assays of developmental fusion behavior. Given the etiology and prevalence of cleft palate and the relatively simple architecture and composition of the embryonic palate, we sought to develop a three-dimensional culture system that mimics the embryonic palate and could be used to study fusion behavior in vitro using human cells. We engineered size-controlled human Wharton’s Jelly stromal cell (HWJSC) spheroids and established that 7 days of culture in osteogenesis differentiation medium was sufficient to promote an osteogenic phenotype consistent with embryonic palatal mesenchyme. HWJSC spheroids supported the attachment of human epidermal keratinocyte progenitor cells (HPEKp) on the outer spheroid surface likely through deposition of collagens I and IV, fibronectin, and laminin by mesenchymal spheroids. HWJSC spheroids coated in HPEKp cells exhibited fusion behavior in culture, as indicated by the removal of epithelial cells from the seams between spheroids, that was dependent on epidermal growth factor signaling and fibroblast growth factor signaling in agreement with palate fusion literature. The method described here may broadly apply to the generation of three-dimensional epithelial-mesenchymal co-cultures to study developmental fusion events in a format that is amenable to predictive toxicology applications.
Collapse
Affiliation(s)
- David G. Belair
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Cynthia J. Wolf
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Carmen Wood
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Hongzu Ren
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Rachel Grindstaff
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - William Padgett
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Adam Swank
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Denise MacMillan
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Anna Fisher
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Witold Winnik
- Research Cores Unit, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
| | - Barbara D. Abbott
- Toxicity Assessment Division, US EPA, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
36
|
van de Peppel J, Strini T, Tilburg J, Westerhoff H, van Wijnen AJ, van Leeuwen JP. Identification of Three Early Phases of Cell-Fate Determination during Osteogenic and Adipogenic Differentiation by Transcription Factor Dynamics. Stem Cell Reports 2017; 8:947-960. [PMID: 28344004 PMCID: PMC5390132 DOI: 10.1016/j.stemcr.2017.02.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 02/20/2017] [Accepted: 02/20/2017] [Indexed: 01/08/2023] Open
Abstract
Age-related skeletal degeneration in patients with osteoporosis is characterized by decreased bone mass and occurs concomitant with an increase in bone marrow adipocytes. Using microarray expression profiling with high temporal resolution, we identified gene regulatory events in early stages of osteogenic and adipogenic lineage commitment of human mesenchymal stromal cells (hMSCs). Data analysis revealed three distinct phases when cells adopt a committed expression phenotype: initiation of differentiation (0-3 hr, phase I), lineage acquisition (6-24 hr, phase II), and early lineage progression (48-96 hr, phase III). Upstream regulator analysis identified 34 transcription factors (TFs) in phase I with a role in hMSC differentiation. Interestingly, expression levels of identified TFs did not always change and indicate additional post-transcriptional regulatory mechanisms. Functional analysis revealed that forced expression of IRF2 enhances osteogenic differentiation. Thus, IRF2 and other early-responder TFs may control osteogenic cell fate of MSCs and should be considered in mechanistic models that clarify bone-anabolic changes during clinical progression of osteoporosis.
Collapse
Affiliation(s)
- Jeroen van de Peppel
- Bone and Calcium Metabolism, Department Internal Medicine, Erasmus MC, Wytemaweg 80, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Tanja Strini
- Bone and Calcium Metabolism, Department Internal Medicine, Erasmus MC, Wytemaweg 80, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Julia Tilburg
- Bone and Calcium Metabolism, Department Internal Medicine, Erasmus MC, Wytemaweg 80, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Hans Westerhoff
- Synthetic Systems Biology, University of Amsterdam, 1081 HZ Amsterdam, the Netherlands; Molecular Cell Physiology, VU University Amsterdam, 1081 HZ Amsterdam, the Netherlands; Systems Biology, MCISB, University of Manchester, Manchester M1 7DN, UK
| | - Andre J van Wijnen
- Bone and Calcium Metabolism, Department Internal Medicine, Erasmus MC, Wytemaweg 80, Postbus 2040, 3000 CA Rotterdam, the Netherlands; Department of Orthopedic Surgery, Biochemistry & Molecular Biology, and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Johannes P van Leeuwen
- Bone and Calcium Metabolism, Department Internal Medicine, Erasmus MC, Wytemaweg 80, Postbus 2040, 3000 CA Rotterdam, the Netherlands.
| |
Collapse
|
37
|
Vitamin D Effects on Osteoblastic Differentiation of Mesenchymal Stem Cells from Dental Tissues. Stem Cells Int 2016; 2016:9150819. [PMID: 27956902 PMCID: PMC5124467 DOI: 10.1155/2016/9150819] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/24/2016] [Accepted: 09/25/2016] [Indexed: 12/22/2022] Open
Abstract
1α,25-Dihydroxyvitamin D3 (1,25(OH)2D3), the active metabolite of vitamin D (Vit D), increases intestinal absorption of calcium and phosphate, maintaining a correct balance of bone remodeling. Vit D has an anabolic effect on the skeletal system and is key in promoting osteoblastic differentiation of human Mesenchymal Stem Cells (hMSCs) from bone marrow. MSCs can be also isolated from the immature form of the tooth, the dental bud: Dental Bud Stem Cells (DBSCs) are adult stem cells that can effectively undergo osteoblastic differentiation. In this work we investigated the effect of Vit D on DBSCs differentiation into osteoblasts. Our data demonstrate that DBSCs, cultured in an opportune osteogenic medium, differentiate into osteoblast-like cells; Vit D treatment stimulates their osteoblastic features, increasing the expression of typical markers of osteoblastogenesis like RUNX2 and Collagen I (Coll I) and, in a more important way, determining a higher production of mineralized matrix nodules.
Collapse
|
38
|
Wang J, Wu P, Chen PC, Lee C, Chen W, Hung S. Generation of Osteosarcomas from a Combination of Rb Silencing and c-Myc Overexpression in Human Mesenchymal Stem Cells. Stem Cells Transl Med 2016; 6:512-526. [PMID: 28191765 PMCID: PMC5442803 DOI: 10.5966/sctm.2015-0226] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 07/15/2016] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma (OS) was a malignant tumor occurring with unknown etiology that made prevention and early diagnosis difficult. Mesenchymal stem cells (MSCs), which were found in bone marrow, were claimed to be a possible origin of OS but with little direct evidence. We aimed to characterize OS cells transformed from human MSCs (hMSCs) and identify their association with human primary OS cells and patient survival. Genetic modification with p53 or retinoblastoma (Rb) knockdown and c-Myc or Ras overexpression was applied for hMSC transformation. Transformed cells were assayed for proliferation, differentiation, tumorigenecity, and gene expression profile. Only the combination of Rb knockdown and c-Myc overexpression successfully transformed hMSCs derived from four individual donors, with increasing cell proliferation, decreasing cell senescence rate, and increasing ability to form colonies and spheres in serum-free medium. These transformed cells lost the expression of certain surface markers, increased in osteogenic potential, and decreased in adipogenic potential. After injection in immunodeficient mice, these cells formed OS-like tumors, as evidenced by radiographic analyses and immunohistochemistry of various OS markers. Microarray with cluster analysis revealed that these transformed cells have gene profiles more similar to patient-derived primary OS cells than their normal MSC counterparts. Most importantly, comparison of OS patient tumor samples revealed that a combination of Rb loss and c-Myc overexpression correlated with a decrease in patient survival. This study successfully transformed human MSCs to OS-like cells by Rb knockdown and c-Myc overexpression that may be a useful platform for further investigation of preventive and target therapy for human OS. Stem Cells Translational Medicine 2017;6:512-526.
Collapse
Affiliation(s)
- Jir‐You Wang
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Department of Orthopaedics, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Institute of Traditional Medicine, School of Medicine, National Yang‐Ming University, Taipei, Taiwan, Republic of China
| | - Po‐Kuei Wu
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Department of Orthopaedics, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Paul Chih‐Hsueh Chen
- Department of Orthopaedics, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Chia‐Wen Lee
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Department of Orthopaedics, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Wei‐Ming Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Department of Orthopaedics, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Shih‐Chieh Hung
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Department of Orthopaedics, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Institute of Traditional Medicine, School of Medicine, National Yang‐Ming University, Taipei, Taiwan, Republic of China
- Institute of Clinical Medicine, School of Medicine, National Yang‐Ming University, Taipei, Taiwan, Republic of China
- Department of Pharmacology, School of Medicine, National Yang‐Ming University, Taipei, Taiwan, Republic of China
- Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
- Integrative Stem Cell Center, China Medical University Hospital, Taichung, Taiwan, Republic of China
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan, Republic of China
| |
Collapse
|
39
|
Electrochemical deposition of mineralized BSA/collagen coating. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 66:66-76. [DOI: 10.1016/j.msec.2016.04.088] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 01/29/2016] [Accepted: 04/24/2016] [Indexed: 01/18/2023]
|
40
|
van Zoelen EJ, Duarte I, Hendriks JM, van der Woning SP. TGFβ-induced switch from adipogenic to osteogenic differentiation of human mesenchymal stem cells: identification of drug targets for prevention of fat cell differentiation. Stem Cell Res Ther 2016; 7:123. [PMID: 27562730 PMCID: PMC5000485 DOI: 10.1186/s13287-016-0375-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 07/12/2016] [Accepted: 07/25/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Patients suffering from osteoporosis show an increased number of adipocytes in their bone marrow, concomitant with a reduction in the pool of human mesenchymal stem cells (hMSCs) that are able to differentiate into osteoblasts, thus leading to suppressed osteogenesis. METHODS In order to be able to interfere with this process, we have investigated in-vitro culture conditions whereby adipogenic differentiation of hMSCs is impaired and osteogenic differentiation is promoted. By means of gene expression microarray analysis, we have investigated genes which are potential targets for prevention of fat cell differentiation. RESULTS Our data show that BMP2 promotes both adipogenic and osteogenic differentiation of hMSCs, while transforming growth factor beta (TGFβ) inhibits differentiation into both lineages. However, when cells are cultured under adipogenic differentiation conditions, which contain cAMP-enhancing agents such as IBMX of PGE2, TGFβ promotes osteogenic differentiation, while at the same time inhibiting adipogenic differentiation. Gene expression and immunoblot analysis indicated that IBMX-induced suppression of HDAC5 levels plays an important role in the inhibitory effect of TGFβ on osteogenic differentiation. By means of gene expression microarray analysis, we have investigated genes which are downregulated by TGFβ under adipogenic differentiation conditions and may therefore be potential targets for prevention of fat cell differentiation. We thus identified nine genes for which FDA-approved drugs are available. Our results show that drugs directed against the nuclear hormone receptor PPARG, the metalloproteinase ADAMTS5, and the aldo-keto reductase AKR1B10 inhibit adipogenic differentiation in a dose-dependent manner, although in contrast to TGFβ they do not appear to promote osteogenic differentiation. CONCLUSIONS The approach chosen in this study has resulted in the identification of new targets for inhibition of fat cell differentiation, which may not only be relevant for prevention of osteoporosis, but also of obesity.
Collapse
Affiliation(s)
- Everardus J van Zoelen
- Department of Cell and Applied Biology, Faculty of Science, Radboud University Nijmegen, PO Box 9010, 6500 GL, Nijmegen, The Netherlands. .,Present Address: Department of Cell and Applied Biology, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands.
| | - Isabel Duarte
- Department of Cell and Applied Biology, Faculty of Science, Radboud University Nijmegen, PO Box 9010, 6500 GL, Nijmegen, The Netherlands.,Present Address: Department of Cell and Applied Biology, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands.,Present Address: Systems Biology and Bioinformatics Laboratory (SysBioLab), University of Algarve, Faro, Portugal
| | - José M Hendriks
- Department of Cell and Applied Biology, Faculty of Science, Radboud University Nijmegen, PO Box 9010, 6500 GL, Nijmegen, The Netherlands.,Present Address: Department of Cell and Applied Biology, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands.,Present Address: Department of Physical Organic Chemistry, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Sebastian P van der Woning
- Department of Cell and Applied Biology, Faculty of Science, Radboud University Nijmegen, PO Box 9010, 6500 GL, Nijmegen, The Netherlands.,Present Address: Department of Cell and Applied Biology, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands.,Present Address: ARGENX BVBA, Technologiepark 30, B-9052, Zwijnaarde, Belgium
| |
Collapse
|
41
|
Zhuang J, Lin J, Li J, Weng W, Cheng K, Wang H. Alternating potentials assisted electrochemical deposition of mineralized collagen coatings. Colloids Surf B Biointerfaces 2015; 136:479-87. [DOI: 10.1016/j.colsurfb.2015.09.050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 08/24/2015] [Accepted: 09/26/2015] [Indexed: 11/30/2022]
|
42
|
Mechanism of Action of Bortezomib and the New Proteasome Inhibitors on Myeloma Cells and the Bone Microenvironment: Impact on Myeloma-Induced Alterations of Bone Remodeling. BIOMED RESEARCH INTERNATIONAL 2015; 2015:172458. [PMID: 26579531 PMCID: PMC4633537 DOI: 10.1155/2015/172458] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/22/2015] [Accepted: 09/27/2015] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM) is characterized by a high capacity to induce alterations in the bone remodeling process. The increase in osteoclastogenesis and the suppression of osteoblast formation are both involved in the pathophysiology of the bone lesions in MM. The proteasome inhibitor (PI) bortezomib is the first drug designed and approved for the treatment of MM patients by targeting the proteasome. However, recently novel PIs have been developed to overcome bortezomib resistance. Interestingly, several preclinical data indicate that the proteasome complex is involved in both osteoclast and osteoblast formation. It is also evident that bortezomib either inhibits osteoclast differentiation induced by the receptor activator of nuclear factor kappa B (NF-κB) ligand (RANKL) or stimulates the osteoblast differentiation. Similarly, the new PIs including carfilzomib and ixazomib can inhibit bone resorption and stimulate the osteoblast differentiation. In a clinical setting, PIs restore the abnormal bone remodeling by normalizing the levels of bone turnover markers. In addition, a bone anabolic effect was described in responding MM patients treated with PIs, as demonstrated by the increase in the osteoblast number. This review summarizes the preclinical and clinical evidence on the effects of bortezomib and other new PIs on myeloma bone disease.
Collapse
|
43
|
Bionaz M, Monaco E, Wheeler MB. Transcription Adaptation during In Vitro Adipogenesis and Osteogenesis of Porcine Mesenchymal Stem Cells: Dynamics of Pathways, Biological Processes, Up-Stream Regulators, and Gene Networks. PLoS One 2015; 10:e0137644. [PMID: 26398344 PMCID: PMC4580618 DOI: 10.1371/journal.pone.0137644] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 07/27/2015] [Indexed: 12/20/2022] Open
Abstract
The importance of mesenchymal stem cells (MSC) for bone regeneration is growing. Among MSC the bone marrow-derived stem cells (BMSC) are considered the gold standard in tissue engineering and regenerative medicine; however, the adipose-derived stem cells (ASC) have very similar properties and some advantages to be considered a good alternative to BMSC. The molecular mechanisms driving adipogenesis are relatively well-known but mechanisms driving osteogenesis are poorly known, particularly in pig. In the present study we have used transcriptome analysis to unravel pathways and biological functions driving in vitro adipogenesis and osteogenesis in BMSC and ASC. The analysis was performed using the novel Dynamic Impact Approach and functional enrichment analysis. In addition, a k-mean cluster analysis in association with enrichment analysis, networks reconstruction, and transcription factors overlapping analysis were performed in order to uncover the coordination of biological functions underlining differentiations. Analysis indicated a larger and more coordinated transcriptomic adaptation during adipogenesis compared to osteogenesis, with a larger induction of metabolism, particularly lipid synthesis (mostly triglycerides), and a larger use of amino acids for synthesis of feed-forward adipogenic compounds, larger cell signaling, lower cell-to-cell interactions, particularly for the cytoskeleton organization and cell junctions, and lower cell proliferation. The coordination of adipogenesis was mostly driven by Peroxisome Proliferator-activated Receptors together with other known adipogenic transcription factors. Only a few pathways and functions were more induced during osteogenesis compared to adipogenesis and some were more inhibited during osteogenesis, such as cholesterol and protein synthesis. Up-stream transcription factor analysis indicated activation of several lipid-related transcription regulators (e.g., PPARs and CEBPα) during adipogenesis but osteogenesis was driven by inhibition of several up-stream regulators, such as MYC. Between MSCs the data indicated an ‘adipocyte memory’ in ASC with also an apparent lower immunogenicity compared to BMSC during differentiations. Overall the analysis allowed proposing a dynamic model for the adipogenic and osteogenic differentiation in porcine ASC and BMSC.
Collapse
Affiliation(s)
- Massimo Bionaz
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Elisa Monaco
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Matthew B. Wheeler
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
44
|
Jeong Y, Swami S, Krishnan AV, Williams JD, Martin S, Horst RL, Albertelli MA, Feldman BJ, Feldman D, Diehn M. Inhibition of Mouse Breast Tumor-Initiating Cells by Calcitriol and Dietary Vitamin D. Mol Cancer Ther 2015; 14:1951-61. [PMID: 25934710 DOI: 10.1158/1535-7163.mct-15-0066] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/28/2015] [Indexed: 12/29/2022]
Abstract
The anticancer actions of vitamin D and its hormonally active form, calcitriol, have been extensively documented in clinical and preclinical studies. However, the mechanisms underlying these actions have not been completely elucidated. Here, we examined the effect of dietary vitamin D and calcitriol on mouse breast tumor-initiating cells (TICs, also known as cancer stem cells). We focused on MMTV-Wnt1 mammary tumors, for which markers for isolating TICs have previously been validated. We confirmed that these tumors expressed functional vitamin D receptors and estrogen receptors (ER) and exhibited calcitriol-induced molecular responses including ER downregulation. Following orthotopic implantation of MMTV-Wnt1 mammary tumor cells into mice, calcitriol injections or a vitamin D-supplemented diet caused a striking delay in tumor appearance and growth, whereas a vitamin D-deficient diet accelerated tumor appearance and growth. Calcitriol inhibited TIC tumor spheroid formation in a dose-dependent manner in primary cultures and inhibited TIC self-renewal in secondary passages. A combination of calcitriol and ionizing radiation inhibited spheroid formation more than either treatment alone. Further, calcitriol significantly decreased TIC frequency as evaluated by in vivo limiting dilution analyses. Calcitriol inhibition of TIC spheroid formation could be overcome by the overexpression of β-catenin, suggesting that the inhibition of Wnt/β-catenin pathway is an important mechanism mediating the TIC inhibitory activity of calcitriol in this tumor model. Our findings indicate that vitamin D compounds target breast TICs reducing tumor-initiating activity. Our data also suggest that combining vitamin D compounds with standard therapies may enhance anticancer activity and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Youngtae Jeong
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Srilatha Swami
- Department of Medicine, Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, California
| | - Aruna V Krishnan
- Department of Medicine, Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, California
| | - Jasmaine D Williams
- Department of Pediatrics/Endocrinology, Stanford University School of Medicine, Stanford, California. Cancer Biology Program, Stanford University School of Medicine, Stanford, California
| | - Shanique Martin
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Megan A Albertelli
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California
| | - Brian J Feldman
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California. Department of Pediatrics/Endocrinology, Stanford University School of Medicine, Stanford, California. Cancer Biology Program, Stanford University School of Medicine, Stanford, California
| | - David Feldman
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California. Department of Medicine, Division of Endocrinology, Gerontology, and Metabolism, Stanford University School of Medicine, Stanford, California.
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California. Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
45
|
Satué M, Ramis JM, Monjo M. UV-activated 7-dehydrocholesterol-coated titanium implants promote differentiation of human umbilical cord mesenchymal stem cells into osteoblasts. J Biomater Appl 2015; 30:770-9. [DOI: 10.1177/0885328215582324] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Vitamin D metabolites are essential for bone regeneration and mineral homeostasis. The vitamin D precursor 7-dehydrocholesterol can be used after UV irradiation to locally produce active vitamin D by osteoblastic cells. Furthermore, UV-irradiated 7-dehydrocholesterol is a biocompatible coating for titanium implants with positive effects on osteoblast differentiation. In this study, we examined the impact of titanium implants surfaces coated with UV-irradiated 7-dehydrocholesterol on the osteogenic differentiation of human umbilical cord mesenchymal stem cells. First, the synthesis of cholecalciferol (D3) was achieved through the incubation of the UV-activated 7-dehydrocholesterol coating for 48 h at 23℃. Further, we investigated in vitro the biocompatibility of this coating in human umbilical cord mesenchymal stem cells and its potential to enhance their differentiation towards the osteogenic lineage. Human umbilical cord mesenchymal stem cells cultured onto UV-irradiated 7-dehydrocholesterol-coated titanium implants surfaces, combined with osteogenic supplements, upregulated the gene expression of several osteogenic markers and showed higher alkaline phosphatase activity and calcein blue staining, suggesting increased mineralization. Thus, our results show that the use of UV irradiation on 7-dehydrocholesterol -treated titanium implants surfaces generates a bioactive coating that promotes the osteogenic differentiation of human umbilical cord mesenchymal stem cells, with regenerative potential for improving osseointegration in titanium-based bone anchored implants.
Collapse
Affiliation(s)
- María Satué
- Department of Fundamental Biology and Health Sciences, Research Institute on Health Sciences (IUNICS), University of Balearic Islands, Palma de Mallorca, Spain; Instituto de Investigación Sanitaria de Palma, Palma de Mallorca, Spain
| | - Joana M Ramis
- Department of Fundamental Biology and Health Sciences, Research Institute on Health Sciences (IUNICS), University of Balearic Islands, Palma de Mallorca, Spain; Instituto de Investigación Sanitaria de Palma, Palma de Mallorca, Spain
| | - Marta Monjo
- Department of Fundamental Biology and Health Sciences, Research Institute on Health Sciences (IUNICS), University of Balearic Islands, Palma de Mallorca, Spain; Instituto de Investigación Sanitaria de Palma, Palma de Mallorca, Spain
| |
Collapse
|
46
|
Wu S, Liu X, Gao C. Role of adsorbed proteins on hydroxyapatite-coated titanium in osteoblast adhesion and osteogenic differentiation. Sci Bull (Beijing) 2015. [DOI: 10.1007/s11434-015-0753-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Lysdahl H, Baatrup A, Foldager CB, Bünger C. Preconditioning Human Mesenchymal Stem Cells with a Low Concentration of BMP2 Stimulates Proliferation and Osteogenic Differentiation In Vitro. Biores Open Access 2014; 3:278-85. [PMID: 25469313 PMCID: PMC4245882 DOI: 10.1089/biores.2014.0044] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Clinical trials using bone morphogenetic protein-2 (BMP2) for bone reconstruction have shown promising results. However, the relatively high concentration needed to be effective raises concerns for efficacy and safety. The aim of this study was to investigate the osteogenic effect of an alternative treatment strategy in which human bone marrow–derived mesenchymal stem cells (hMSCs) are preconditioned with low concentrations of BMP2 for a short time in vitro. hMSCs in suspension were stimulated for 15 min with 10 and 20 ng/mL of BMP2. After the BMP2 was removed, the cells were seeded and cultured in osteogenic medium. The effects of preconditioning were analyzed with regard to proliferation and expression of osteogenic markers at both gene and protein level. The results were compared to those from cultures with continuous BMP2 stimulation. A significant increase in proliferation was seen with both precondition and continuous stimulation with BMP2, with no difference between the treatments. Preconditioning with BMP2 significantly increased gene expression of RUNX2, COLI, ALP, and OC, and protein levels of COLI and ALP. This was not found with continuous stimulation. The role of preconditioning with BMP2 in osteogenesis was validated by findings of increased gene expression of SMAD1 and an increase in dual phosphorylation of ser 463 and ser 465 in the SMAD 1/5/8 pathway. We concluded that preconditioning hMSCs with BMP2 stimulates osteogenesis: proliferation with matrix secretion and matrix maturation of hMSCs. This implies that preconditioning with BMP2 might be more effective at inducing proliferation and osteogenic differentiation of hMSCs than continuous stimulation. Preconditioning with BMP2 could benefit the clinical application of BMP2 since side effects from high-dose treatments could be avoided.
Collapse
Affiliation(s)
- Helle Lysdahl
- Orthopaedic Research Laboratory, Aarhus University Hospital , Aarhus, Denmark
| | - Anette Baatrup
- Orthopaedic Research Laboratory, Aarhus University Hospital , Aarhus, Denmark
| | | | - Cody Bünger
- Orthopaedic Research Laboratory, Aarhus University Hospital , Aarhus, Denmark
| |
Collapse
|
48
|
He M, Xu M, Zhang B, Liang J, Chen P, Lee JY, Johnson TA, Li H, Yang X, Dai J, Liang L, Gui L, Qi Q, Huang J, Li Y, Adair LS, Aung T, Cai Q, Cheng CY, Cho MC, Cho YS, Chu M, Cui B, Gao YT, Go MJ, Gu D, Gu W, Guo H, Hao Y, Hong J, Hu Z, Hu Y, Huang J, Hwang JY, Ikram MK, Jin G, Kang DH, Khor CC, Kim BJ, Kim HT, Kubo M, Lee J, Lee J, Lee NR, Li R, Li J, Liu J, Longe J, Lu W, Lu X, Miao X, Okada Y, Ong RTH, Qiu G, Seielstad M, Sim X, Song H, Takeuchi F, Tanaka T, Taylor PR, Wang L, Wang W, Wang Y, Wu C, Wu Y, Xiang YB, Yamamoto K, Yang H, Liao M, Yokota M, Young T, Zhang X, Kato N, Wang QK, Zheng W, Hu FB, Lin D, Shen H, Teo YY, Mo Z, Wong TY, Lin X, Mohlke KL, Ning G, Tsunoda T, Han BG, Shu XO, Tai ES, Wu T, Qi L. Meta-analysis of genome-wide association studies of adult height in East Asians identifies 17 novel loci. Hum Mol Genet 2014; 24:1791-800. [PMID: 25429064 DOI: 10.1093/hmg/ddu583] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Human height is associated with risk of multiple diseases and is profoundly determined by an individual's genetic makeup and shows a high degree of ethnic heterogeneity. Large-scale genome-wide association (GWA) analyses of adult height in Europeans have identified nearly 180 genetic loci. A recent study showed high replicability of results from Europeans-based GWA studies in Asians; however, population-specific loci may exist due to distinct linkage disequilibrium patterns. We carried out a GWA meta-analysis in 93 926 individuals from East Asia. We identified 98 loci, including 17 novel and 81 previously reported loci, associated with height at P < 5 × 10(-8), together explaining 8.89% of phenotypic variance. Among the newly identified variants, 10 are commonly distributed (minor allele frequency, MAF > 5%) in Europeans, with comparable frequencies with in Asians, and 7 single-nucleotide polymorphisms are with low frequency (MAF < 5%) in Europeans. In addition, our data suggest that novel biological pathway such as the protein tyrosine phosphatase family is involved in regulation of height. The findings from this study considerably expand our knowledge of the genetic architecture of human height in Asians.
Collapse
Affiliation(s)
- Meian He
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Hubei, Wuhan 430030, China,
| | - Min Xu
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115, USA, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism and
| | - Ben Zhang
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jun Liang
- Department of Endocrinology, The Central Hospital of Xuzhou, Affiliated Hospital of Southeast University, Xuzhou, Jiangsu 221009, China
| | - Peng Chen
- Saw Swee Hock School of Public Health
| | - Jong-Young Lee
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do 363-700, Republic of Korea
| | | | - Huaixing Li
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaobo Yang
- Institute of Urology and Nephrology, First Affiliated Hospital & Center for Genomic and Personalized Medicine, Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Juncheng Dai
- Department of Epidemiology & Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Liming Liang
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115, USA
| | - Lixuan Gui
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Hubei, Wuhan 430030, China
| | - Qibin Qi
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115, USA
| | - Jinyan Huang
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115, USA
| | - Yanping Li
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115, USA
| | | | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore 168751
| | - Qiuyin Cai
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Ching-Yu Cheng
- Saw Swee Hock School of Public Health, Department of Ophthalmology, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore 168751, Duke-National University of Singapore Graduate Medical School, Singapore, Singapore 169857
| | - Myeong-Chan Cho
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do 363-700, Republic of Korea
| | - Yoon Shin Cho
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do 363-700, Republic of Korea
| | - Minjie Chu
- Department of Epidemiology & Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Bin Cui
- Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism and
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Min Jin Go
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do 363-700, Republic of Korea
| | - Dongfeng Gu
- State Key Laboratory of Cardiovascular Disease, Department of Evidence Based Medicine, Fuwai Hospital, National Center of Cardiovascular Diseases
| | - Weiqiong Gu
- Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism and
| | - Huan Guo
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Hubei, Wuhan 430030, China
| | - Yongchen Hao
- State Key Laboratory of Cardiovascular Disease, Department of Evidence Based Medicine, Fuwai Hospital, National Center of Cardiovascular Diseases
| | - Jie Hong
- Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism and
| | - Zhibin Hu
- Department of Epidemiology & Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | | | - Jianfeng Huang
- State Key Laboratory of Cardiovascular Disease, Department of Evidence Based Medicine, Fuwai Hospital, National Center of Cardiovascular Diseases
| | - Joo-Yeon Hwang
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do 363-700, Republic of Korea
| | - Mohammad Kamran Ikram
- Saw Swee Hock School of Public Health, Department of Ophthalmology, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore 168751, Department of Ophthalmology, Erasmus Medical Center, Rotterdam 3015, The Netherlands, Memory Aging & Cognition Centre, National University Health System, Singapore, Singapore 119228
| | - Guangfu Jin
- Department of Epidemiology & Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Dae-Hee Kang
- Department of Preventive Medicine, Seoul National University, College of Medicine, Seoul 110-799, Republic of Korea
| | - Chiea Chuen Khor
- Saw Swee Hock School of Public Health, Department of Ophthalmology, Department of Paediatrics National University Health Systems, Agency for Science, Technology and Research, Genome Institute of Singapore, Singapore, Singapore 138672
| | - Bong-Jo Kim
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do 363-700, Republic of Korea
| | - Hung Tae Kim
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do 363-700, Republic of Korea
| | | | | | - Juyoung Lee
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do 363-700, Republic of Korea
| | - Nanette R Lee
- Office of Population Studies Foundation, University of San Carlos, Cebu City 6000, Philippines
| | - Ruoying Li
- Department of Medicine, Yong Loo Lin School of Medicine
| | - Jun Li
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Hubei, Wuhan 430030, China
| | - JianJun Liu
- Saw Swee Hock School of Public Health, Agency for Science, Technology and Research, Genome Institute of Singapore, Singapore, Singapore 138672
| | - Jirong Longe
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Wei Lu
- Shanghai Municipal Center for Disease Control & Prevention, 1380 Zhong Shan Road (W), Shanghai 200336, China
| | - Xiangfeng Lu
- State Key Laboratory of Cardiovascular Disease, Department of Evidence Based Medicine, Fuwai Hospital, National Center of Cardiovascular Diseases
| | - Xiaoping Miao
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Hubei, Wuhan 430030, China
| | | | | | - Gaokun Qiu
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Hubei, Wuhan 430030, China
| | - Mark Seielstad
- Department of Paediatrics National University Health Systems
| | - Xueling Sim
- Saw Swee Hock School of Public Health, Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109-2029, USA
| | - Huaidong Song
- State Key Laboratory of Medical Genomics, Molecular Medical Center, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Toshihiro Tanaka
- Laboratory for Cardiovascular Diseases, RIKEN Center for Genomic Medicine, Yokohama 230-0045, Japan
| | - Phil R Taylor
- Division of Cancer Epidemiology & Genetics, Genetic Epidemiology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Laiyuan Wang
- State Key Laboratory of Cardiovascular Disease, Department of Evidence Based Medicine, Fuwai Hospital, National Center of Cardiovascular Diseases
| | - Weiqing Wang
- Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism and
| | - Yiqin Wang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chen Wu
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ying Wu
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yong-Bing Xiang
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ken Yamamoto
- Department of Molecular Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Handong Yang
- Department of Cardiology, Dongfeng Central Hospital, Dongfeng Motor Corporation and Hubei University of Medicine, Shiyan, Hubei 442008, China
| | - Ming Liao
- Institute of Urology and Nephrology, First Affiliated Hospital & Center for Genomic and Personalized Medicine
| | - Mitsuhiro Yokota
- Department of Genome Science, Aichi-Gakuin University, School of Dentistry, Nagoya 464-8650, Japan
| | - Terri Young
- Duke-National University of Singapore Graduate Medical School, Singapore, Singapore 169857, Duke Eye Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Xiaomin Zhang
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Hubei, Wuhan 430030, China
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Qing K Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Institute, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Wei Zheng
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Frank B Hu
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115, USA
| | - Dongxin Lin
- State Key Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Hongbing Shen
- Department of Epidemiology & Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Yik Ying Teo
- Saw Swee Hock School of Public Health, NUS Graduate School for Integrative Science and Engineering, Life Sciences Institute, Department of Statistics and Applied Probability, National University of Singapore, Singapore, Singapore 119077, Agency for Science, Technology and Research, Genome Institute of Singapore, Singapore, Singapore 138672
| | - Zengnan Mo
- Institute of Urology and Nephrology, First Affiliated Hospital & Center for Genomic and Personalized Medicine
| | - Tien Yin Wong
- Department of Ophthalmology, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore 168751
| | - Xu Lin
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Guang Ning
- Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism and
| | | | - Bok-Ghee Han
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do 363-700, Republic of Korea
| | - Xiao-Ou Shu
- Vanderbilt Epidemiology Center, Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, Department of Medicine, Yong Loo Lin School of Medicine, Duke-National University of Singapore Graduate Medical School, Singapore, Singapore 169857
| | - Tangchun Wu
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Hubei, Wuhan 430030, China
| | - Lu Qi
- Department of Nutrition, Harvard School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
49
|
Roson-Burgo B, Sanchez-Guijo F, Del Cañizo C, De Las Rivas J. Transcriptomic portrait of human Mesenchymal Stromal/Stem Cells isolated from bone marrow and placenta. BMC Genomics 2014; 15:910. [PMID: 25326687 PMCID: PMC4287589 DOI: 10.1186/1471-2164-15-910] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 09/24/2014] [Indexed: 02/08/2023] Open
Abstract
Background Human Mesenchymal Stromal/Stem Cells (MSCs) are adult multipotent cells that behave in a highly plastic manner, inhabiting the stroma of several tissues. The potential utility of MSCs is nowadays strongly investigated in the field of regenerative medicine and cell therapy, although many questions about their molecular identity remain uncertain. Results MSC primary cultures from human bone marrow (BM) and placenta (PL) were derived and verified by their immunophenotype standard pattern and trilineage differentiation potential. Then, a broad characterization of the transcriptome of these MSCs was performed using RNA deep sequencing (RNA-Seq). Quantitative analysis of these data rendered an extensive expression footprint that includes 5,271 protein-coding genes. Flow cytometry assays of canonical MSC CD-markers were congruent with their expression levels detected by the RNA-Seq. Expression of other recently proposed MSC markers (CD146, Nestin and CD271) was tested in the placenta samples, finding only CD146 and Nestin. Functional analysis revealed enrichment in stem cell related genes and mesenchymal regulatory transcription factors (TFs). Analysis of TF binding sites (TFBSs) identified 11 meta-regulators, including factors KLF4 and MYC among them. Epigenetically, hypomethylated promoter patterns supported the active expression of the MSC TFs found. An interaction network of these TFs was built to show up their links and relations. Assessment of dissimilarities between cell origins (BM versus PL) disclosed two hundred differentially expressed genes enrolled in microenvironment processes related to the cellular niche, as regulation of bone formation and blood vessel morphogenesis for the case of BM-MSCs. By contrast genes overexpressed in PL-MSCs showed functional enrichment on mitosis, negative regulation of cell-death and embryonic morphogenesis that supported the higher growth rates observed in the cultures of these fetal cells and their closer links with development processes. Conclusions The results present a transcriptomic portrait of the human MSCs isolated from bone marrow and placenta. The data are released as a cell-specific resource, providing a comprehensive expression footprint of the MSCs useful to better understand their cellular and molecular biology and for further investigations on the isolation and biomedical use of these multipotent cells. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-910) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (IBMCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Cientificas (CSIC), Salamanca, Spain.
| |
Collapse
|
50
|
Abstract
Fat and bone have a complicated relationship. Although obesity has been associated with low fracture risk, there is increasing evidence that some of the factors that are released by peripheral fat into the circulation may also have a deleterious effect on bone mass, thus, predisposing to fractures. More importantly, the local interaction between fat and bone within the bone marrow seems to play a significant role in the pathogenesis of age-related bone loss and osteoporosis. This "local interaction" occurs inside the bone marrow and is associated with the autocrine and paracrine release of fatty acids and adipokines, which affect the cells in their vicinity including the osteoblasts, reducing their function and survival. In this review, we explore the particularities of the fat and bone cell interactions within the bone marrow, their significance in the pathogenesis of osteoporosis, and the potential therapeutic applications that regulating marrow fat may have in the near future as a novel pharmacologic treatment for osteoporosis.
Collapse
Affiliation(s)
- Sandra Bermeo
- Ageing Bone Research Program, Sydney Medical School Nepean, The University of Sydney, Level 5, South Block, Nepean Hospital, Penrith, NSW., Australia, 2750
| | | | | |
Collapse
|