1
|
Qiao X, Yang Y, Zhao Y, Wu X, Zhang L, Cai X, Ji J, Boström KI, Yao Y. Aurora Kinase A Regulates Cell Transitions in Glucocorticoid-Induced Bone Loss. Cells 2023; 12:2434. [PMID: 37887278 PMCID: PMC10605378 DOI: 10.3390/cells12202434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/22/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
Glucocorticoid-induced bone loss is a severe and toxic effect of long-term therapy with glucocorticoids, which are currently prescribed for millions of people worldwide. Previous studies have uncovered that glucocorticoids reciprocally converted osteoblast lineage cells into endothelial-like cells to cause bone loss and showed that the modulations of Foxc2 and Osterix were the causative factors that drove this harmful transition of osteoblast lineage cells. Here, we find that the inhibition of aurora kinase A halts this transition and prevents glucocorticoid-induced bone loss. We find that aurora A interacts with the glucocorticoid receptor and show that this interaction is required for glucocorticoids to modulate Foxc2 and Osterix. Together, we identify a new potential approach to counteracting unwanted transitions of osteoblast lineage cells in glucocorticoid treatment and may provide a novel strategy for ameliorating glucocorticoid-induced bone loss.
Collapse
Affiliation(s)
- Xiaojing Qiao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA
| | - Yang Yang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA
| | - Yan Zhao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA
| | - Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA
| | - Li Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA
| | - Xinjiang Cai
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA
| | - Jaden Ji
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA
| | - Kristina I. Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA
- The Molecular Biology Institute at UCLA, Los Angeles, CA 90095-1570, USA
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1679, USA
| |
Collapse
|
2
|
Qiao X, Wu X, Zhao Y, Yang Y, Zhang L, Cai X, Ma JA, Ji J, Lyons K, Boström KI, Yao Y. Cell Transitions Contribute to Glucocorticoid-Induced Bone Loss. Cells 2023; 12:1810. [PMID: 37508475 PMCID: PMC10377921 DOI: 10.3390/cells12141810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/26/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Glucocorticoid-induced bone loss is a toxic effect of long-term therapy with glucocorticoids resulting in a significant increase in the risk of fracture. Here, we find that glucocorticoids reciprocally convert osteoblast-lineage cells into endothelial-like cells. This is confirmed by lineage tracing showing the induction of endothelial markers in osteoblast-lineage cells following glucocorticoid treatment. Functional studies show that osteoblast-lineage cells isolated from glucocorticoid-treated mice lose their capacity for bone formation but simultaneously improve vascular repair. We find that the glucocorticoid receptor directly targets Foxc2 and Osterix, and the modulations of Foxc2 and Osterix drive the transition of osteoblast-lineage cells to endothelial-like cells. Together, the results suggest that glucocorticoids suppress osteogenic capacity and cause bone loss at least in part through previously unrecognized osteoblast-endothelial transitions.
Collapse
Affiliation(s)
- Xiaojing Qiao
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Yan Zhao
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Yang Yang
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Li Zhang
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Xinjiang Cai
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Jocelyn A Ma
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Jaden Ji
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Karen Lyons
- Department of Molecular, Cell & Developmental Biology at UCLA, Los Angeles, CA 90095, USA
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- The Molecular Biology Institute at UCLA, Los Angeles, CA 90095, USA
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Neels JG, Gollentz C, Chinetti G. Macrophage death in atherosclerosis: potential role in calcification. Front Immunol 2023; 14:1215612. [PMID: 37469518 PMCID: PMC10352763 DOI: 10.3389/fimmu.2023.1215612] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023] Open
Abstract
Cell death is an important aspect of atherosclerotic plaque development. Insufficient efferocytosis of death cells by phagocytic macrophages leads to the buildup of a necrotic core that impacts stability of the plaque. Furthermore, in the presence of calcium and phosphate, apoptotic bodies resulting from death cells can act as nucleation sites for the formation of calcium phosphate crystals, mostly in the form of hydroxyapatite, which leads to calcification of the atherosclerotic plaque, further impacting plaque stability. Excessive uptake of cholesterol-loaded oxidized LDL particles by macrophages present in atherosclerotic plaques leads to foam cell formation, which not only reduces their efferocytosis capacity, but also can induce apoptosis in these cells. The resulting apoptotic bodies can contribute to calcification of the atherosclerotic plaque. Moreover, other forms of macrophage cell death, such as pyroptosis, necroptosis, parthanatos, and ferroptosis can also contribute by similar mechanisms to plaque calcification. This review focuses on macrophage death in atherosclerosis, and its potential role in calcification. Reducing macrophage cell death and/or increasing their efferocytosis capacity could be a novel therapeutic strategy to reduce the formation of a necrotic core and calcification and thereby improving atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Jaap G. Neels
- Université Côte d’Azur, Institut national de la santé et de la recherche médicale (INSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Claire Gollentz
- Université Côte d’Azur, Centre Hospitalier Universitaire (CHU), Institut national de la santé et de la recherche médicale (NSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Giulia Chinetti
- Université Côte d’Azur, Centre Hospitalier Universitaire (CHU), Institut national de la santé et de la recherche médicale (NSERM), Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| |
Collapse
|
4
|
Huang Y, Wan S, Yang M. Circ_0067680 expedites the osteogenic differentiation of human bone marrow-derived mesenchymal stem cells through miR-4429/CTNNB1/Wnt/β-catenin pathway. Biol Direct 2021; 16:16. [PMID: 34649595 PMCID: PMC8515698 DOI: 10.1186/s13062-021-00302-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/14/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Human bone marrow-derived mesenchymal stem cells (hBMSCs) are the primary source of osteoblasts in vivo. Emerging literatures have unveiled that circular RNAs (circRNAs) are actively drawn in the osteogenic differentiation of mesenchymal stem cells (MSCs). This research mainly illuminated the role of circ_0067680 as well as its regulatory mechanism in osteoblastic differentiation. METHODS In this study, RT-qPCR was to measure the expression of circ_0067680. Functional assays were implemented to assess the role of circ_0067680 in osteogenic differentiation. Besides, RNA pull down, RIP and luciferase reporter assays were carried out to investigate the regulatory mechanism of circ_0067680. RESULTS Circ_0067680, which derived from its host gene divergent protein kinase domain 2A (C3orf58), was up-regulated during osteogenic differentiation of hBMSCs. Besides, circ_0067680 deficiency impeded the osteoblastic differentiation of hBMSCs. Moreover, circ_0067680 served as a ceRNA via sequestering miR-4429 to regulate the expression of catenin beta 1 (CTNNB1), thereby activating the Wnt/β-catenin signaling pathway. CONCLUSION Circ_0067680 accelerated hBMSCs osteogenic differentiation by the miR-4429/CTNNB1/Wnt/β-catenin signaling, which might be used as a potential biomarker for osteoblastic differentiation.
Collapse
Affiliation(s)
- Yuansheng Huang
- Traumatic Orthopedics, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Su Wan
- Department of Gynecology, Yijishan Hospital of Wannan Medical College, No. 2, Zheshan West Road, Wuhu, 241001, Anhui, China.
| | - Min Yang
- Traumatic Orthopedics, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| |
Collapse
|
5
|
Chinetti G, Neels JG. Roles of Nuclear Receptors in Vascular Calcification. Int J Mol Sci 2021; 22:6491. [PMID: 34204304 PMCID: PMC8235358 DOI: 10.3390/ijms22126491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
Vascular calcification is defined as an inappropriate accumulation of calcium depots occurring in soft tissues, including the vascular wall. Growing evidence suggests that vascular calcification is an actively regulated process, sharing similar mechanisms with bone formation, implicating both inhibitory and inducible factors, mediated by osteoclast-like and osteoblast-like cells, respectively. This process, which occurs in nearly all the arterial beds and in both the medial and intimal layers, mainly involves vascular smooth muscle cells. In the vascular wall, calcification can have different clinical consequences, depending on the pattern, localization and nature of calcium deposition. Nuclear receptors are transcription factors widely expressed, activated by specific ligands that control the expression of target genes involved in a multitude of pathophysiological processes, including metabolism, cancer, inflammation and cell differentiation. Some of them act as drug targets. In this review we describe and discuss the role of different nuclear receptors in the control of vascular calcification.
Collapse
Affiliation(s)
- Giulia Chinetti
- Université Côte d’Azur, CHU, INSERM, C3M, 06204 Nice, France;
| | - Jaap G. Neels
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France
| |
Collapse
|
6
|
Jeon HH, Teixeira H, Tsai A. Mechanistic Insight into Orthodontic Tooth Movement Based on Animal Studies: A Critical Review. J Clin Med 2021; 10:jcm10081733. [PMID: 33923725 PMCID: PMC8072633 DOI: 10.3390/jcm10081733] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 01/09/2023] Open
Abstract
Alveolar bone remodeling in orthodontic tooth movement (OTM) is a highly regulated process that coordinates bone resorption by osteoclasts and new bone formation by osteoblasts. Mechanisms involved in OTM include mechano-sensing, sterile inflammation-mediated osteoclastogenesis on the compression side and tensile force-induced osteogenesis on the tension side. Several intracellular signaling pathways and mechanosensors including the cilia and ion channels transduce mechanical force into biochemical signals that stimulate formation of osteoclasts or osteoblasts. To date, many studies were performed in vitro or using human gingival crevicular fluid samples. Thus, the use of transgenic animals is very helpful in examining a cause and effect relationship. Key cell types that participate in mediating the response to OTM include periodontal ligament fibroblasts, mesenchymal stem cells, osteoblasts, osteocytes, and osteoclasts. Intercellular signals that stimulate cellular processes needed for orthodontic tooth movement include receptor activator of nuclear factor-κB ligand (RANKL), tumor necrosis factor-α (TNF-α), dickkopf Wnt signaling pathway inhibitor 1 (DKK1), sclerostin, transforming growth factor beta (TGF-β), and bone morphogenetic proteins (BMPs). In this review, we critically summarize the current OTM studies using transgenic animal models in order to provide mechanistic insight into the cellular events and the molecular regulation of OTM.
Collapse
|
7
|
Xie J, Lou Q, Zeng Y, Liang Y, Xie S, Xu Q, Yuan L, Wang J, Jiang L, Mou L, Lin D, Zhao M. Single-Cell Atlas Reveals Fatty Acid Metabolites Regulate the Functional Heterogeneity of Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:653308. [PMID: 33912565 PMCID: PMC8075002 DOI: 10.3389/fcell.2021.653308] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/09/2021] [Indexed: 12/28/2022] Open
Abstract
Bone marrow mesenchymal stem cells (MSCs) are widely used clinically due to their versatile roles in multipotency, immunomodulation, and hematopoietic stem cell (HSC) niche function. However, cellular heterogeneity limits MSCs in the consistency and efficacy of their clinical applications. Metabolism regulates stem cell function and fate decision; however, how metabolites regulate the functional heterogeneity of MSCs remains elusive. Here, using single-cell RNA sequencing, we discovered that fatty acid pathways are involved in the regulation of lineage commitment and functional heterogeneity of MSCs. Functional assays showed that a fatty acid metabolite, butyrate, suppressed the self-renewal, adipogenesis, and osteogenesis differentiation potential of MSCs with increased apoptosis. Conversely, butyrate supplement significantly promoted HSC niche factor expression in MSCs, which suggests that butyrate supplement may provide a therapeutic approach to enhance their HSC niche function. Overall, our work demonstrates that metabolites are essential to regulate the functional heterogeneity of MSCs.
Collapse
Affiliation(s)
- Jiayi Xie
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qi Lou
- Shenzhen Lansi Institute of Artificial Intelligence in Medicine, Shenzhen, China.,The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yunxin Zeng
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yingying Liang
- Shenzhen Lansi Institute of Artificial Intelligence in Medicine, Shenzhen, China.,The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| | - Siyu Xie
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Quanhui Xu
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Lisha Yuan
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Jin Wang
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Linjia Jiang
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lisha Mou
- Shenzhen Lansi Institute of Artificial Intelligence in Medicine, Shenzhen, China
| | - Dongjun Lin
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Meng Zhao
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Shenzhen Lansi Institute of Artificial Intelligence in Medicine, Shenzhen, China.,Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| |
Collapse
|
8
|
Aghlmandi A, Nikshad A, Safaralizadeh R, Warkiani ME, Aghebati-Maleki L, Yousefi M. Microfluidics as efficient technology for the isolation and characterization of stem cells. EXCLI JOURNAL 2021; 20:426-443. [PMID: 33746671 PMCID: PMC7975637 DOI: 10.17179/excli2020-3028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/15/2021] [Indexed: 01/09/2023]
Abstract
The recent years have been passed with significant progressions in the utilization of microfluidic technologies for cellular investigations. The aim of microfluidics is to mimic small-scale body environment with features like optical transparency. Microfluidics can screen and monitor different cell types during culture and study cell function in response to stimuli in a fully controlled environment. No matter how the microfluidic environment is similar to in vivo environment, it is not possible to fully investigate stem cells behavior in response to stimuli during cell proliferation and differentiation. Researchers have used stem cells in different fields from fundamental researches to clinical applications. Many cells in the body possess particular functions, but stem cells do not have a specific task and can turn into almost any type of cells. Stem cells are undifferentiated cells with the ability of changing into specific cells that can be essential for the body. Researchers and physicians are interested in stem cells to use them in testing the function of the body's systems and solving their complications. This review discusses the recent advances in utilizing microfluidic techniques for the analysis of stem cells, and mentions the advantages and disadvantages of using microfluidic technology for stem cell research.
Collapse
Affiliation(s)
- Afsoon Aghlmandi
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Aylin Nikshad
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Majid Ebrahimi Warkiani
- The School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | | | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
| |
Collapse
|
9
|
Effect of 20(S)-Hydroxycholesterol on Multilineage Differentiation of Mesenchymal Stem Cells Isolated from Compact Bones in Chicken. Genes (Basel) 2020; 11:genes11111360. [PMID: 33213081 PMCID: PMC7698591 DOI: 10.3390/genes11111360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/17/2022] Open
Abstract
Bone health and body weight gain have significant economic and welfare importance in the poultry industry. Mesenchymal stem cells (MSCs) are common progenitors of different cell lineages such as osteoblasts, adipocytes, and myocytes. Specific oxysterols have shown to be pro-osteogenic and anti-adipogenic in mouse and human MSCs. To determine the effect of 20(S)-hydroxycholesterol (20S) on osteogenic, adipogenic, and myogenic differentiation in chicken, mesenchymal stem cells isolated from compact bones of broiler chickens (cBMSCs) were subjected to various doses of 20S, and markers of lineage-specific mRNA were analyzed using real-time PCR and cell cytochemistry. Further studies were conducted to evaluate the molecular mechanisms involved in lineage-specific differentiation pathways. Like human and mouse MSCs, 20S oxysterol expressed pro-osteogenic, pro-myogenic, and anti-adipogenic differentiation potential in cBMSCs. Moreover, 20(S)-Hydroxycholesterol induced markers of osteogenic genes and myogenic regulatory factors when exposed to cBMSCs treated with their specific medium. In contrast, 20S oxysterol suppressed expression of adipogenic marker genes when exposed to cBMSCs treated with OA, an adipogenic precursor of cBMSCs. To elucidate the molecular mechanism by which 20S exerts its differentiation potential in all three lineages, we focused on the hedgehog signaling pathway. The hedgehog inhibitor, cyclopamine, completely reversed the effect of 20S induced expression of osteogenic and anti-adipogenic mRNA. However, there was no change in the mRNA expression of myogenic genes. The results showed that 20S oxysterol promotes osteogenic and myogenic differentiation and decreases adipocyte differentiation of cBMSCs. This study also showed that the induction of osteogenesis and adipogenesis inhibition in cBMSCs by 20S is mediated through the hedgehog signaling mechanism. The results indicated that 20(S) could play an important role in the differentiation of chicken-derived MSCs and provided the theory basis on developing an intervention strategy to regulate skeletal, myogenic, and adipogenic differentiation in chicken, which will contribute to improving chicken bone health and meat quality. The current results provide the rationale for the further study of regulatory mechanisms of bioactive molecules on the differentiation of MSCs in chicken, which can help to address skeletal health problems in poultry.
Collapse
|
10
|
Three-Dimensional Culture System of Cancer Cells Combined with Biomaterials for Drug Screening. Cancers (Basel) 2020; 12:cancers12102754. [PMID: 32987868 PMCID: PMC7601447 DOI: 10.3390/cancers12102754] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary For the research and development of drug discovery, it is of prime importance to construct the three-dimensional (3D) tissue models in vitro. To this end, the enhancement design of cell function and activity by making use of biomaterials is essential. In this review, 3D culture systems of cancer cells combined with several biomaterials for anticancer drug screening are introduced. Abstract Anticancer drug screening is one of the most important research and development processes to develop new drugs for cancer treatment. However, there is a problem resulting in gaps between the in vitro drug screening and preclinical or clinical study. This is mainly because the condition of cancer cell culture is quite different from that in vivo. As a trial to mimic the in vivo cancer environment, there has been some research on a three-dimensional (3D) culture system by making use of biomaterials. The 3D culture technologies enable us to give cancer cells an in vitro environment close to the in vivo condition. Cancer cells modified to replicate the in vivo cancer environment will promote the biological research or drug discovery of cancers. This review introduces the in vitro research of 3D cell culture systems with biomaterials in addition to a brief summary of the cancer environment.
Collapse
|
11
|
Butein Promotes Lineage Commitment of Bone Marrow-Derived Stem Cells into Osteoblasts via Modulating ERK1/2 Signaling Pathways. Molecules 2020; 25:molecules25081885. [PMID: 32325749 PMCID: PMC7221720 DOI: 10.3390/molecules25081885] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/13/2020] [Accepted: 04/17/2020] [Indexed: 01/16/2023] Open
Abstract
Butein is a phytochemical that belongs to the chalcone family of flavonoids and has antitumor, anti-inflammatory, and anti-osteoclastic bone resorption activities. This study aims to investigate the effects of butein on the differentiation potential of mouse primary bone marrow-derived mesenchymal stem cells (mBMSCs) into osteoblast and adipocyte lineages. Primary cultures of mBMSCs are treated with different doses of butein during its differentiation. Osteoblast differentiation is assessed by alkaline phosphatase (ALP) activity quantification and Alizarin red staining for matrix mineralization, while adipogenesis is assessed by quantification of lipid accumulation using Oil Red O staining. Osteoblastic and adipocytic gene expression markers are determined by quantitative real-time PCR (qPCR). Western blot analysis is used to study the activation of extracellular signal-regulated kinase (ERK1/2). Interestingly, butein promotes the lineage commitment of mBMSCs into osteoblasts, while suppressing their differentiation into adipocytes in a dose-dependent manner. A similar effect of butein is confirmed in human (h) primary BMSCs. Occurring at the molecular level, butein significantly upregulates the mRNA expression of osteoblast-related genes, while downregulating the expression of adipocyte-related genes. The mechanism of butein-induced osteogenesis is found to be mediated by activating the ERK1/2 signaling pathway. To conclude, we identify butein as a novel nutraceutical compound with an osteo-anabolic activity to promote the lineage commitment of BMSCs into osteoblast versus adipocyte. Thus, butein can be a plausible therapeutic drug for enhancing bone formation in osteoporotic patients.
Collapse
|
12
|
Hu F, Zhao Y, Hui Z, Xing F, Yang J, Lee I, Zhang X, Pan L, Xu J. Regulation of intracellular Ca2+/CaMKII signaling by TRPV4 membrane translocation during osteoblastic differentiation. BIOPHYSICS REPORTS 2019. [DOI: 10.1007/s41048-019-00100-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
AbstractBone constantly remodels between resorption by osteoclasts and formation by osteoblasts; therefore the functions of osteoblasts are pivotal for maintaining homeostasis of bone mass. Transient receptor potential vanilloid 4 (TRPV4), a type of mechanosensitive channel, has been reported to be a key regulator in bone remodeling. However, the relationship between TRPV4 and osteoblast function remains largely elusive. Only little is known about the spatial distribution change of TRPV4 during osteoblastic differentiation and related signal events. Based on three-dimensional super-resolution microscopy, our results clearly showed a different distribution of TRPV4 in undifferentiated and differentiated osteoblasts, which reflected the plasma membrane translocation of TRPV4 along with prolonged differentiation. GSK1016790A (GSK101), the most potent agonist of TRPV4, triggered rapid calcium entry and calmodulin-dependent protein kinase II (CaMKII) phosphorylation via TRPV4 activation in a differentiation-dependent manner, indicating that the abundance of TRPV4 at the cell surface resulting from differentiation may be related to the modulation of Ca2+ response and CaMKII activity. These data provide compelling evidences for the plasma membrane translocation of TRPV4 during osteoblastic differentiation as well as demonstrate the regulation of downstream Ca2+/CaMKII signaling.
Collapse
|
13
|
Morsoleto MJMDS, Sella V, Machado P, Bomfim FD, Fernandes MH, Morgado F, Lopes Filho GDJ, Plapler H. Effect of low power laser in biomodulation of cultured osteoblastic cells of Wistar rats1. Acta Cir Bras 2019; 34:e201900210. [PMID: 30843943 PMCID: PMC6585914 DOI: 10.1590/s0102-8650201900210] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/22/2019] [Indexed: 02/08/2023] Open
Abstract
Purpose To analyze aspects of the biomodulating effect of light in biological
tissues, bone cells from surgical explants of the femur of rats were
irradiated with low intensity laser. Methods Bone cells were cultured and irradiated with LASER light (GaAlAs). Growth,
cell viability, mineralized matrix formation, total protein dosage,
immunostimulatory properties, cytochemical analysis, gene expression of bone
proteins were examined using live cell imaging and cell counting by
colorimetric assay. The gene expression of: alkaline phosphatase (ALP), type
1 collagen, osteocalcin and osteopontin through the real-time polymerase
chain reaction. Results At 8 days, the viability of the irradiated culture was 82.3% and 72.4% in
non-irradiated cells. At 18 days, the cellular viability (with laser) was
77.42% and 47.62% without laser. At 8 days, the total protein concentration
was 21.622 mg / mol in the irradiated group and 16, 604 mg / mol in the
non-irradiated group and at 18 days the concentration was 37.25 mg / mol in
the irradiated group and 24, 95 mg / mol in the non-irradiated group. Conclusion The laser interfered in the histochemical reaction, cell viability, matrix
mineralization, and maintained the cellular expression of proteins
Collapse
Affiliation(s)
- Maria Jose Misael da Silva Morsoleto
- Postdoctoral, Postgraduate Program in Interdisciplinary Surgical Sciences, Universidade Federal de São Paulo (UNIFESP), Brazil. Design, intellectual and scientific content of the study; acquisition and interpretation of data; manuscript preparation and writing
| | - Valeria Sella
- Fellow PhD degree, Postgraduate Program in Interdisciplinary Surgical Science, UNIFESP, Sao Paulo-SP, Brazil. Conception and design of the study
| | - Paula Machado
- Physiotherapist, Postgraduate Program in Interdisciplinary Surgical Sciences, UNIFESP, Sao Paulo-SP, Brazil. Technical procedures
| | - Fernando do Bomfim
- Fellow PhD degree, Postgraduate Program in Interdisciplinary Surgical Sciences, UNIFESP, Sao Paulo-SP, Brazil. Technical procedures
| | - Maria Helena Fernandes
- Associate Professor, Department of Pharmacology and Cellular Compatibility, Dental Medicine Faculty, Universidade do Porto, Portugal. Histopathological examinations, Analysis and interpretation of data
| | - Fernando Morgado
- Associate Professor, Department of Biology, Universidade de Aveiro, Portugal. Analysis and interpretation of data, statistics analysis
| | - Gaspar de Jesus Lopes Filho
- Associate Professor, Department of Surgery, Medical School, UNIFESP, Sao Paulo-SP, Brazil. Critical revision, final approval
| | - Helio Plapler
- Associate Professor, Department of Surgery, Medical School, UNIFESP, Sao Paulo-SP, Brazil. Conception, design, intellectual and scientific content of the study; critical revision
| |
Collapse
|
14
|
Lv ZT, Liang S, Chen K, Zhang JM, Cheng P, Guo JC, Yang Q, Zhou CH, Liao H, Chen AM. FNDC4 Inhibits RANKL-Induced Osteoclast Formation by Suppressing NF- κB Activation and CXCL10 Expression. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3936257. [PMID: 29977911 PMCID: PMC5998196 DOI: 10.1155/2018/3936257] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/18/2018] [Accepted: 04/29/2018] [Indexed: 11/18/2022]
Abstract
FNDC4 acts as an anti-inflammatory factor on macrophages and improves mouse model of induced colitis. Considering osteoclast formation is characterized by the activation of inflammation-related pathways, we thus speculated that FNDC4 may play a pivotal role in this process. RT-qPCR analysis was performed to confirm the expression of osteoclast formation related genes in primary murine bone marrow macrophages (BMMs). RANKL-treated BMMs were cultured with FNDC4 to evaluate the effect of FNDC4 on osteoclast differentiation. TRAP staining and bone resorption pits assay were used to assess osteoclast formation and bone resorption, respectively. Luciferase assay and western blotting analysis were conducted to determine whether FNDC4 inhibited osteoclast formation via NF-κB signaling in RAW 264.7 cells. Furthermore, to identify gene signatures in FNDC4-treated BMMs and to use these to elucidate the underlying molecular mechanisms during osteoclast formation, we adopted a bioinformatics approach by downloading the GSE76172 gene expression profiling dataset from the Gene Expression Omnibus (GEO) database. FNDC4 inhibited RANKL-induced osteoclastogenesis and mature osteoclast resorptive function in a dose-dependent manner. Results of NF-κB luciferase assay suggested that FNDC4 could significantly suppress the RANKL-induced NF-κB transcriptional activity. Based on the protein-protein interaction network, CXCL10 was identified as the differentially expressed gene with the highest connectivity degree (degree = 23); it was drastically downregulated in the presence of FNDC4, but supplementation of CXCL10 (10 ng/mL) partially ameliorated the FNDC4-induced inhibition of osteoclast formation. Taken together, we speculated that FNDC4 could suppress osteoclast formation via NF-κB pathway and downregulation of CXCL10.
Collapse
Affiliation(s)
- Zheng-tao Lv
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuang Liang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kun Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jia-ming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jia-chao Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qing Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chen-he Zhou
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hui Liao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - An-min Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
15
|
Yogui FC, Momesso GAC, Faverani LP, Polo TOB, Ramalho-Ferreira G, Hassumi JS, Rossi AC, Freire AR, Prado FB, Okamoto R. A SERM increasing the expression of the osteoblastogenesis and mineralization-related proteins and improving quality of bone tissue in an experimental model of osteoporosis. J Appl Oral Sci 2018; 26:e20170329. [PMID: 29742257 PMCID: PMC5933824 DOI: 10.1590/1678-7757-2017-0329] [Citation(s) in RCA: 10] [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: 07/25/2017] [Revised: 09/26/2017] [Accepted: 10/09/2017] [Indexed: 01/17/2023] Open
Abstract
Raloxifene is an antiresorptive drug, selective estrogen receptor modulator (SERM) used in the treatment of osteoporosis. Objective To evaluate proteins related to bone repair at the peri-implant bone in a rat model of osteoporosis treated with raloxifene. Material and Methods 72 rats were divided into three groups: SHAM (healthy animals), OVX (ovariectomized animals), and RLX (ovariectomized animals treated with raloxifene). Raloxifene was administered by gavage (1 mg/kg/day). Tibial implantation was performed 30 days after ovariectomy, and animals were euthanized at 14, 42, and 60 days postoperatively. Samples were collected and analyzed by immunohistochemical reactions, molecular analysis, and microtomographic parameters. Results RLX showed intense staining of all investigated proteins at both time points except for RUNX2. These results were similar to SHAM and opposite to OVX, showing mild staining. The PCR gene expression of OC and ALP values for RLX (P<0.05) followed by SHAM and OVX groups. For BSP data, the highest expression was observed in the RLX groups and the lowest expression was observed in the OVX groups (P<0.05). For RUNX2 data, RLX and SHAM groups showed greater values compared to OVX (P<0.05). At 60 days postoperatively, microtomography parameters, related to closed porosity, showed higher values for (Po.N), (Po.V), and (Po) in RLX and SHAM groups, whereas OVX groups showed lower results (P<0.05); (BV) values (P=0.009); regarding total porosity (Po.tot), RLX group had statistically significant lower values than OVX and SHAM groups (P=0.009). Regarding the open porosity (Po.V and Po), the SHAM group presented the highest values, followed by OVX and RLX groups (P<0.05). The Structural Model Index (SMI), RLX group showed a value closer to zero than SHAM group (P<0.05). Conclusions Raloxifene had a positive effect on the expression of osteoblastogenesis/mineralization-related proteins and on micro-CT parameters related to peri-implant bone healing.
Collapse
Affiliation(s)
- Fernanda Costa Yogui
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia de Araçatuba, Departamento de Ciências Básicas, Araçatuba, São Paulo, Brasil
| | - Gustavo Antonio Correa Momesso
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia de Araçatuba, Departamento de Cirurgia e Clínica Integrada, Araçatuba, São Paulo, Brasil
| | - Leonardo Perez Faverani
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia de Araçatuba, Departamento de Cirurgia e Clínica Integrada, Araçatuba, São Paulo, Brasil
| | - Tarik Ocon Braga Polo
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia de Araçatuba, Departamento de Cirurgia e Clínica Integrada, Araçatuba, São Paulo, Brasil
| | - Gabriel Ramalho-Ferreira
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia de Araçatuba, Departamento de Cirurgia e Clínica Integrada, Araçatuba, São Paulo, Brasil
| | - Jaqueline Suemi Hassumi
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia de Araçatuba, Departamento de Cirurgia e Clínica Integrada, Araçatuba, São Paulo, Brasil
| | - Ana Cláudia Rossi
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Departamento de Anatomia, Piracicaba, SP, Brasil
| | - Alexandre Rodrigues Freire
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Departamento de Anatomia, Piracicaba, SP, Brasil
| | - Felippe Bevilacqua Prado
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Departamento de Anatomia, Piracicaba, SP, Brasil
| | - Roberta Okamoto
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia de Araçatuba, Departamento de Ciências Básicas, Araçatuba, São Paulo, Brasil
| |
Collapse
|
16
|
Ribeiro M, Fernandes MH, Beppu MM, Monteiro FJ, Ferraz MP. Silk fibroin/nanohydroxyapatite hydrogels for promoted bioactivity and osteoblastic proliferation and differentiation of human bone marrow stromal cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 89:336-345. [PMID: 29752106 DOI: 10.1016/j.msec.2018.04.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 02/20/2018] [Accepted: 04/15/2018] [Indexed: 02/07/2023]
Abstract
Silk fibroin (SF) is a natural, biocompatible, and biodegradable polymer having a great potential for the successful regeneration of damaged bone tissue. In the present work, nanohydroxyapatite (nanoHA) was incorporated into SF polymer to form a bioactive composite hydrogel for applications as bone implants. The degradation and bioactive properties of SF/nanoHA composite hydrogels were evaluated. Additionally, biological investigations of human bone marrow stromal cells (hBMSCs) viability, proliferation and differentiation to the osteoblastic phenotype were conducted. The incorporation of nanoHA in SF polymer matrices improved the bioactivity of the hydrogels. The biological results highlighted that the SF/nanoHA composite hydrogels are suitable for hBMSCs attachment and proliferation, while a test for alkaline phosphatase (ALP) and bone morphogenetic protein 2 (BMP-2) expression suggested osteoblast differentiation. Additionally, a cell staining method for ALP allowed to observe cell infiltration with active production of ALP by the infiltrated cells, paving the way to use the proposed composite hydrogel for bone tissue regeneration.
Collapse
Affiliation(s)
- Marta Ribeiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto 4150-180, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, Departamento de Engenharia Metalúrgica e Materiais, Porto 4200-465, Portugal.
| | - Maria H Fernandes
- Laboratory for Bone Metabolism and Regeneration, Faculdade de Medicina Dentária, Universidade do Porto, Porto 4200-393, Portugal; REQUIMTE/LAQV - U. Porto, Porto, Portugal
| | - Marisa M Beppu
- School of Chemical Engineering, University of Campinas, Campinas, SP 13083-852, Brazil
| | - Fernando J Monteiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto 4150-180, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, Departamento de Engenharia Metalúrgica e Materiais, Porto 4200-465, Portugal
| | - Maria P Ferraz
- FP-ENAS/CEBIMED - University Fernando Pessoa Energy, Environment and Health Research Unit/Biomedical Research Center, Porto 4249-004, Portugal
| |
Collapse
|
17
|
Olimpio RMC, de Oliveira M, De Sibio MT, Moretto FCF, Deprá IC, Mathias LS, Gonçalves BM, Rodrigues BM, Tilli HP, Coscrato VE, Costa SMB, Mazeto GMFS, Fernandes CJC, Zambuzzi WF, Saraiva PP, Maria DA, Nogueira CR. Cell viability assessed in a reproducible model of human osteoblasts derived from human adipose-derived stem cells. PLoS One 2018; 13:e0194847. [PMID: 29641603 PMCID: PMC5895002 DOI: 10.1371/journal.pone.0194847] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/09/2018] [Indexed: 01/04/2023] Open
Abstract
Human adipose tissue-derived stem cells (hASCs) have been subjected to extensive investigation because of their self-renewal properties and potential to restore damaged tissues. In the literature, there are several protocols for differentiating hASCs into osteoblasts, but there is no report on the control of cell viability during this process. In this study, we used osteoblasts derived from hASCs of patients undergoing abdominoplasty. The cells were observed at the beginning and end of bone matrix formation, and the expression of proteins involved in this process, including alkaline phosphatase and osteocalcin, was assessed. RANKL, Osterix, Runx2, Collagen3A1, Osteopontin and BSP expression levels were analyzed using real-time PCR, in addition to a quantitative assessment of protein levels of the markers CD45, CD105, STRO-1, and Nanog, using immunofluorescence. Rhodamine (Rho123), cytochrome-c, caspase-3, P-27, cyclin D1, and autophagy cell markers were analyzed by flow cytometry to demonstrate potential cellular activity and the absence of apoptotic and tumor cell processes before and after cell differentiation. The formation of bone matrix, along with calcium nodules, was observed after 16 days of osteoinduction. The gene expression levels of RANKL, Osterix, Runx2, Collagen3A1, Osteopontin, BSP and alkaline phosphatase activity were also elevated after 16 days of osteoinduction, whereas the level of osteocalcin was higher after 21 days of osteoinduction. Our data also showed that the cells had a high mitochondrial membrane potential and a low expression of apoptotic and tumor markers, both before and after differentiation. Cells were viable after the different phases of differentiation. This proposed methodology, using markers to evaluate cell viability, is therefore successful in assessing different phases of stem cell isolation and differentiation.
Collapse
Affiliation(s)
- Regiane M. C. Olimpio
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
- * E-mail:
| | - Miriane de Oliveira
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Maria T. De Sibio
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Fernanda C. F. Moretto
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Igor C. Deprá
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Lucas S. Mathias
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Bianca M. Gonçalves
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Bruna M. Rodrigues
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Helena P. Tilli
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Virgínia E. Coscrato
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Sarah M. B. Costa
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Gláucia M. F. S. Mazeto
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Célio J. C. Fernandes
- Institute of Biosciences, Department of Chemistry and Biochemistry, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Willian F. Zambuzzi
- Institute of Biosciences, Department of Chemistry and Biochemistry, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Patrícia P. Saraiva
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Durvanei A. Maria
- Biochemistry and Biophysics Laboratory, Butantan Institute, São Paulo, São Paulo, Brazil
| | - Célia R. Nogueira
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| |
Collapse
|
18
|
Rocca A, Tafuri D, Paccone M, Giuliani A, Zamboli AGI, Surfaro G, Paccone A, Compagna R, Amato M, Serra R, Amato B. Cell Based Therapeutic Approach in Vascular Surgery: Application and Review. Open Med (Wars) 2017; 12:308-322. [PMID: 29071303 PMCID: PMC5651406 DOI: 10.1515/med-2017-0045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 08/16/2017] [Indexed: 01/14/2023] Open
Abstract
Multipotent stem cells - such as mesenchymal stem/stromal cells and stem cells derived from different sources like vascular wall are intensely studied to try to rapidly translate their discovered features from bench to bedside. Vascular wall resident stem cells recruitment, differentiation, survival, proliferation, growth factor production, and signaling pathways transduced were analyzed. We studied biological properties of vascular resident stem cells and explored the relationship from several factors as Matrix Metalloproteinases (MMPs) and regulations of biological, translational and clinical features of these cells. In this review we described a translational and clinical approach to Adult Vascular Wall Resident Multipotent Vascular Stem Cells (VW-SCs) and reported their involvement in alternative clinical approach as cells based therapy in vascular disease like arterial aneurysms or peripheral arterial obstructive disease.
Collapse
Affiliation(s)
- Aldo Rocca
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, ItalyVia Sergio Pansini, 80131Naples, Italy
| | - Domenico Tafuri
- Department of Sport Sciences and Wellness, University of Naples “Parthenope”, Naples, Italy
| | - Marianna Paccone
- Department of Medicine and Health Sciences Vincenzo Tiberio, University of Molise, Campobasso, Italy
| | - Antonio Giuliani
- A.O.R.N. A. Cardarelli Hepatobiliary and Liver Transplatation Center, Naples, Italy
| | | | - Giuseppe Surfaro
- Antonio Cardarelli Hospital, General Surgery Unit, Campobasso, Italy
| | - Andrea Paccone
- Department of Medicine and Health Sciences Vincenzo Tiberio, University of Molise, Campobasso, Italy
| | - Rita Compagna
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Maurizo Amato
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, University of Catanzaro, Catanzaro, Italy
| | - Bruno Amato
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
19
|
Chinetti-Gbaguidi G, Daoudi M, Rosa M, Vinod M, Louvet L, Copin C, Fanchon M, Vanhoutte J, Derudas B, Belloy L, Haulon S, Zawadzki C, Susen S, Massy ZA, Eeckhoute J, Staels B. Human Alternative Macrophages Populate Calcified Areas of Atherosclerotic Lesions and Display Impaired RANKL-Induced Osteoclastic Bone Resorption Activity. Circ Res 2017; 121:19-30. [DOI: 10.1161/circresaha.116.310262] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 11/16/2022]
Abstract
Rationale:
Vascular calcification is a process similar to bone formation leading to an inappropriate deposition of calcium phosphate minerals in advanced atherosclerotic plaques. Monocyte-derived macrophages, located in atherosclerotic lesions and presenting heterogeneous phenotypes, from classical proinflammatory M1 to alternative anti-inflammatory M2 macrophages, could potentially display osteoclast-like functions.
Objective:
To characterize the phenotype of macrophages located in areas surrounding the calcium deposits in human atherosclerotic plaques.
Methods and Results:
Macrophages near calcium deposits display an alternative phenotype being both CD68 and mannose receptor–positive, expressing carbonic anhydrase type II, but relatively low levels of cathepsin K. In vitro interleukin-4-polarization of human primary monocytes into macrophages results in lower expression and activity of cathepsin K compared with resting unpolarized macrophages. Moreover, interleukin-4 polarization lowers expression levels of the osteoclast transcriptional activator nuclear factor of activated T cells type c-1, associated with increased gene promoter levels of the transcriptional repression mark H3K27me3 (histone 3 lysine 27 trimethylation). Despite higher expression of the receptor activator of nuclear factor κB receptor, receptor activator of nuclear factor κB ligand/macrophage colony-stimulating factor induction of nuclear factor of activated T cells type c-1 and cathepsin K expression is defective in these macrophages because of reduced Erk/c-fos–mediated downstream signaling resulting in impaired bone resorption capacity.
Conclusions:
These results indicate that macrophages surrounding calcium deposits in human atherosclerotic plaques are phenotypically defective being unable to resorb calcification.
Collapse
Affiliation(s)
- Giulia Chinetti-Gbaguidi
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Mehdi Daoudi
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Mickael Rosa
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Manjula Vinod
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Loïc Louvet
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Corinne Copin
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Mélanie Fanchon
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Jonathan Vanhoutte
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Bruno Derudas
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Loic Belloy
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Stephan Haulon
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Christophe Zawadzki
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Sophie Susen
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Ziad A. Massy
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Jérôme Eeckhoute
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| | - Bart Staels
- From the Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011, EGID, Lille, France (G.C.-G., M.D., M.R., M.V., C.C., M.F., J.V., B.D., L.B., C.Z., S.S., J.E., B.S.); University of Côte d’Azur, CHU, Inserm, CNRS, IRCAN, Nice, France (G.C.-G.); Inserm U1088, University of Picardie Jules Verne, and Amiens University Hospital, Amiens, France (L.L.); CHU Lille, Lille, France (S.H.); Division of Nephrology, Ambroise Paré University Hospital, AP-HP, Boulogne-Billancourt (Z.A.M.); and
| |
Collapse
|
20
|
Gambari L, Lisignoli G, Gabusi E, Manferdini C, Paolella F, Piacentini A, Grassi F. Distinctive expression pattern of cystathionine-β-synthase and cystathionine-γ-lyase identifies mesenchymal stromal cells transition to mineralizing osteoblasts. J Cell Physiol 2017; 232:3574-3585. [PMID: 28121025 DOI: 10.1002/jcp.25825] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/20/2016] [Accepted: 01/24/2017] [Indexed: 12/29/2022]
Abstract
Mesenchymal stromal cells (MSCs) are key players in the repair or regeneration of the damaged bone tissue. However, heterogeneity exists between MSCs derived from different donors in their bone formation ability both in vitro and in vivo. The identification of markers defining MSCs with different functional phenotypes is fundamental to maximize their clinical potential. In our previous in vivo study, impaired expression in MSCs of cystathionine-β-synthase (CBS) and cystathionine-γ-lyase (CSE), the two key enzymes in the catabolic pathway of homocysteine, was associated to decreased bone formation and to the onset of osteoporosis in mice. Here, we investigated whether osteogenic differentiation of human MSCs (hMSCs) modulates the expression of CBS and CSE. The expression of CBS and CSE was also assessed during chondrogenesis to confirm the specificity of their expression during osteogenesis. hMSCs displayed a heterogeneous mineralizing capacity between donors (70% of the samples mineralized, while 30% did not mineralize). Inducible expression of CBS and CSE was found to be associated with a mineralizing phenotype in hMSCs. In particular, up-regulation of CSE was restricted to hMSCs undergoing mineralization. During chondrogenesis, CBS was significantly up-regulated while CSE expression was not affected. Ex-vivo findings confirmed that mature h-osteoblasts (hOBs) show consistently higher expression of CBS and CSE than hMSCs. Our data provide the first evidence that the expression of CBS and CSE in hMSCs closely correlates with the transition of hMSCs toward the osteoblastic phenotype and that CSE may constitute a novel marker of osteogenic differentiation.
Collapse
Affiliation(s)
- Laura Gambari
- Laboratorio RAMSES, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Gina Lisignoli
- S.C. Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elena Gabusi
- S.C. Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Cristina Manferdini
- S.C. Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesca Paolella
- S.C. Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Anna Piacentini
- S.C. Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituto Ortopedico Rizzoli, Bologna, Italy
| | | |
Collapse
|
21
|
Wrobel E, Leszczynska J, Brzoska E. The Characteristics Of Human Bone-Derived Cells (HBDCS) during osteogenesis in vitro. Cell Mol Biol Lett 2016; 21:26. [PMID: 28536628 PMCID: PMC5415846 DOI: 10.1186/s11658-016-0027-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 10/28/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The primary human bone-derived cell culture technique is used as a model to study human osteogenesis. Compared to cell line cultures, primary osteoprogenitor and osteoblast cultures provide more complex information about osteogenesis, bone remodeling and regeneration than cell line cultures. METHODS In this study, we isolated human bone-derived cells (HBDCs) and promoted their differentiation into osteoblasts. The following parameters were evaluated: cell number and viability, total protein expression, alkaline phosphatase activity, collagenous matrix production and osteogenic genes expression, i.e., gene coding for type I collagen and alkaline phosphatase. RESULTS It was proved the results show that HBDCs intensively proliferate during the first 7 days of culture followed by differentiation accompanied by an increase in alkaline phosphatase activity. Moreover, it was observed that during the differentiation of HBDCs, the expression of integrin β1 increased. CONCLUSIONS The process was also accompanied by changes in cell shape and rearrangement of the actin cytoskeleton and focal contacts containing FAK and the integrin β1 subunit. We suggest that the β1 integrin subunit may be a suitable new target in studies of the differentiation of primary human osteoblasts in culture.
Collapse
Affiliation(s)
- Edyta Wrobel
- Department of Biophysics and Human Physiology, Faculty of Health Sciences, Medical University of Warsaw, ul. Chalubinskiego 5, 02-004, Warsaw, Poland
| | - Joanna Leszczynska
- Department of Biophysics and Human Physiology, Faculty of Health Sciences, Medical University of Warsaw, ul. Chalubinskiego 5, 02-004, Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, ul. Miecznikowa 1, 02-096, Warsaw, Poland
| |
Collapse
|
22
|
Cha JK, Lim HC, Lee JS, Choi SH, Woo KM, Jung UW. Sinus augmentation using a histone deacetylase inhibitor in a calcium sulfate carrier in rabbit: A pilot study. J Biomed Mater Res B Appl Biomater 2016; 105:1916-1923. [DOI: 10.1002/jbm.b.33727] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 05/13/2016] [Accepted: 05/24/2016] [Indexed: 01/15/2023]
Affiliation(s)
- Jae Kook Cha
- Department of Periodontology; Research Institute for Periodontal Regeneration, Yonsei University, College of Dentistry; Seoul Republic of Korea
| | - Hyun-Chang Lim
- Department of Periodontology; School of Dentistry; Kyung Hee University; Seoul Republic of Korea
| | - Jung-Seok Lee
- Department of Periodontology; Research Institute for Periodontal Regeneration, Yonsei University, College of Dentistry; Seoul Republic of Korea
| | - Seong-Ho Choi
- Department of Periodontology; Research Institute for Periodontal Regeneration, Yonsei University, College of Dentistry; Seoul Republic of Korea
| | - Kyung Mi Woo
- Department of Pharmacology and Dental Therapeutics; School of Dentistry; Seoul National University; Seoul Republic of Korea
| | - Ui-Won Jung
- Department of Periodontology; Research Institute for Periodontal Regeneration, Yonsei University, College of Dentistry; Seoul Republic of Korea
| |
Collapse
|
23
|
Alves EG, Serakides R, Boeloni JN, Rosado IR, Ocarino NM, Oliveira HP, Góes AM, Rezende CM. Estudo comparativo da diferenciação osteogênica das células tronco mesenquimais da medula óssea e do tecido adiposo de cães adultos. PESQUISA VETERINARIA BRASILEIRA 2016. [DOI: 10.1590/s0100-736x201600130004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Resumo: O objetivo deste estudo foi comparar o potencial osteogênico das células tronco mesenquimais extraídas da medula óssea (CTM-MO) com as do tecido adiposo (CTM-AD) de cães adultos. As células foram caracterizadas fenotipicamente quanto à expressão de CD29, CD90, CD34 e CD45 e submetidas à diferenciação adipogênica e condrogênica por 21 dias e osteogênica por 7, 14 e 21 dias. Foram constituídos quatro grupos: 1) CTM-MO em meio osteogênico, 2) CTM-MO em meio basal, 3) CTM-AD em meio osteogênico e 4) CTM-AD em meio basal. Aos 7, 14 e 21 dias de diferenciação osteogênica as culturas foram submetidas às avaliações da conversão de MTT em formazan, da atividade da fosfatase alcalina (FA), da síntese de colágeno e de matriz mineralizada, avaliação do número de células por campo e foram quantificados os transcritos gênicos para osterix, sialoproteina óssea (BSP), osteonectina (ON) e osteocalcina (OC). Tanto as células extraídas da medula óssea quanto do tecido adiposo mostraram elevada expressão de marcadores para células tronco e baixa expressão de marcadores de células hematopoiéticas (menor que 2%). Além disso, foram capazes de se diferenciar em osteoblastos, condrócitos e adipócitos. As CTM-AD submetidas à diferenciação osteogênica mostraram maior conversão do MTT em formazan que as CTM-MO, sob mesmas condições aos 7 e 21 dias. O número de células por campo, a atividade da FA, a síntese de colágeno e de matriz mineralizada foram superior nas CTM-AD em diferenciação, em relação às CTM-MO sob as mesmas condições, em todos os tempos estudados. As expressões de osterix, BSP e OC foram predominantemente superiores nas CTM-MO diferenciadas, mas a expressão de ON foi superior nas CTM-AD diferenciadas aos 7, 14 e 21 dias. Conclui-se que as CTM-AD apresentam maior potencial osteogênico que as CTM-MO quando extraídas de cães adultos.
Collapse
|
24
|
Saeed H, Ahsan M, Saleem Z, Iqtedar M, Islam M, Danish Z, Khan AM. Mesenchymal stem cells (MSCs) as skeletal therapeutics - an update. J Biomed Sci 2016; 23:41. [PMID: 27084089 PMCID: PMC4833928 DOI: 10.1186/s12929-016-0254-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 04/03/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells hold the promise to treat not only several congenital and acquired bone degenerative diseases but also to repair and regenerate morbid bone tissues. Utilizing MSCs, several lines of evidences advocate promising clinical outcomes in skeletal diseases and skeletal tissue repair/regeneration. In this context, both, autologous and allogeneic cell transfer options have been utilized. Studies suggest that MSCs are transplanted either alone by mixing with autogenous plasma/serum or by loading onto repair/induction supportive resorb-able scaffolds. Thus, this review is aimed at highlighting a wide range of pertinent clinical therapeutic options of MSCs in the treatment of skeletal diseases and skeletal tissue regeneration. Additionally, in skeletal disease and regenerative sections, only the early and more recent preclinical evidences are discussed followed by all the pertinent clinical studies. Moreover, germane post transplant therapeutic mechanisms afforded by MSCs have also been conversed. Nonetheless, assertive use of MSCs in the clinic for skeletal disorders and repair is far from a mature therapeutic option, therefore, posed challenges and future directions are also discussed. Importantly, for uniformity at all instances, term MSCs is used throughout the review.
Collapse
Affiliation(s)
- Hamid Saeed
- Section of Clinical Pharmacy, University College of Pharmacy, University of the Punjab, Allama Iqbal Campus, 54000, Lahore, Pakistan.
| | - Muhammad Ahsan
- Section of Clinical Pharmacy, University College of Pharmacy, University of the Punjab, Allama Iqbal Campus, 54000, Lahore, Pakistan
| | - Zikria Saleem
- Section of Clinical Pharmacy, University College of Pharmacy, University of the Punjab, Allama Iqbal Campus, 54000, Lahore, Pakistan
| | - Mehwish Iqtedar
- Department of Bio-technology, Lahore College for Women University, Lahore, Pakistan
| | - Muhammad Islam
- Section of Clinical Pharmacy, University College of Pharmacy, University of the Punjab, Allama Iqbal Campus, 54000, Lahore, Pakistan
| | - Zeeshan Danish
- Section of Clinical Pharmacy, University College of Pharmacy, University of the Punjab, Allama Iqbal Campus, 54000, Lahore, Pakistan
| | - Asif Manzoor Khan
- Department of Biochemistry and Molecular Biology, University of the Southern Denmark, 5230, Odense, Denmark
| |
Collapse
|
25
|
Sawada S, Chosa N, Takizawa N, Yokota J, Igarashi Y, Tomoda K, Kondo H, Yaegashi T, Ishisaki A. Establishment of mesenchymal stem cell lines derived from the bone marrow of green fluorescent protein-transgenic mice exhibiting a diversity in intracellular transforming growth factor-β and bone morphogenetic protein signaling. Mol Med Rep 2016; 13:2023-31. [PMID: 26781600 PMCID: PMC4768972 DOI: 10.3892/mmr.2016.4794] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 08/04/2015] [Indexed: 12/28/2022] Open
Abstract
Cytokines and their intercellular signals regulate the multipotency of mesenchymal stem cells (MSCs). The present study established the MSC lines SG-2, -3, and -5 from the bone marrow of green fluorescent protein (GFP)-transgenic mice. These cell lines clearly expressed mouse MSC markers Sca-1 and CD44, and SG-2 and -5 cells retained the potential for osteogenic and adipogenic differentiation in the absence of members of the transforming growth factor (TGF)-β superfamily. By contrast, SG-3 cells only retained adipogenic differentiation potential. Analysis of cytokine and cytokine receptor expression in these SG cell lines showed that bone morphogenetic protein (BMP) receptor 1B was most highly expressed in the SG-3 cells, which underwent osteogenesis in response to BMP, while TGF-β receptor II was most highly expressed in SG-3 and -5 cells. However, it was unexpectedly noted that phosphorylation of Smad 2, a major transcription factor, was induced by TGF-β1 in SG-2 cells but not in SG-3 or -5 cells. Furthermore, TGF-β1 clearly induced the expression of Smad-interacting transcription factor CCAAT/enhancer binding protein-β in SG-2 but not in SG-3 or -5 cells. These results demonstrated the establishment of TGF-β-responsive SG-2 MSCs, BMP-responsive SG-3 MSCs and TGF-β/BMP-unresponsive SG-5 MSCs, each of which was able to be traced by GFP fluorescence after transplantation into in vivo experimental models. In conclusion, the present study suggested that these cell lines may be used to explore how the TGF-β superfamily affects the proliferation and differentiation status of MSCs in vivo.
Collapse
Affiliation(s)
- Shunsuke Sawada
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028‑3694, Japan
| | - Naoyuki Chosa
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028‑3694, Japan
| | - Naoki Takizawa
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028‑3694, Japan
| | - Jun Yokota
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028‑3694, Japan
| | - Yasuyuki Igarashi
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028‑3694, Japan
| | - Koichi Tomoda
- Department of Otolaryngology, Dentistry and Oral Surgery, Kansai Medical University, Hirakata, Osaka 573‑1010, Japan
| | - Hisatomo Kondo
- Department of Prosthodontics and Oral Implantology, Iwate Medical University School of Dentistry, Morioka, Iwate 020‑8505, Japan
| | - Takashi Yaegashi
- Division of Periodontology, Department of Conservative Dentistry, Iwate Medical University School of Dentistry, Morioka, Iwate 020‑8505, Japan
| | - Akira Ishisaki
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028‑3694, Japan
| |
Collapse
|
26
|
Andrographolide Inhibits Ovariectomy-Induced Bone Loss via the Suppression of RANKL Signaling Pathways. Int J Mol Sci 2015; 16:27470-81. [PMID: 26593901 PMCID: PMC4661897 DOI: 10.3390/ijms161126039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 10/26/2015] [Accepted: 10/26/2015] [Indexed: 11/17/2022] Open
Abstract
Osteoporosis is a debilitating skeletal disorder with an increased risk of low-energy fracture, which commonly occurs among postmenopausal women. Andrographolide (AP), a natural product isolated from Andrographis paniculata, has been found to have anti-inflammatory, anti-cancer, anti-asthmatic, and neuro-protective properties. However, its therapeutic effect on osteoporosis is unknown. In this study, an ovariectomy (OVX) mouse model was used to evaluate the therapeutic effects of AP on post-menopausal osteoporosis by using micro-computed tomography (micro-CT). Bone marrow-derived osteoclast culture was used to examine the inhibitory effect of AP on osteoclastogenesis. Real time PCR was employed to examine the effect of AP on the expression of osteoclast marker genes. The activities of transcriptional factors NF-κB and NFATc1 were evaluated using a luciferase reporter assay, and the IκBα protein level was analyzed by Western blot. We found that OVX mice treated with AP have greater bone volume (BV/TV), trabecular thickness (Tb.Th), and trabecular number (Tb.N) compared to vehicle-treated OVX mice. AP inhibited RANKL-induced osteoclastogenesis, the expression of osteoclast marker genes including cathepsin K (Ctsk), TRACP (Acp5), and NFATc1, as well as the transcriptional activities of NF-κB and NFATc1. In conclusion, our results suggest that AP inhibits estrogen deficiency-induced bone loss in mice via the suppression of RANKL-induced osteoclastogensis and NF-κB and NFATc1 activities and, thus, might have therapeutic potential for osteoporosis.
Collapse
|
27
|
Sakamoto E, Mihara C, Ikuta T, Inagaki Y, Kido J, Nagata T. Inhibitory effects of advanced glycation end-products and Porphyromonas gingivalis
lipopolysaccharide on the expression of osteoblastic markers of rat bone marrow cells in culture. J Periodontal Res 2015. [DOI: 10.1111/jre.12310] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- E. Sakamoto
- Department of Periodontology and Endodontology; Institute of Health Biosciences; Tokushima University Graduate School; Tokushima Japan
| | - C. Mihara
- Department of Periodontology and Endodontology; Institute of Health Biosciences; Tokushima University Graduate School; Tokushima Japan
| | - T. Ikuta
- Department of Periodontology and Endodontology; Institute of Health Biosciences; Tokushima University Graduate School; Tokushima Japan
| | - Y. Inagaki
- Department of Periodontology and Endodontology; Institute of Health Biosciences; Tokushima University Graduate School; Tokushima Japan
| | - J. Kido
- Department of Periodontology and Endodontology; Institute of Health Biosciences; Tokushima University Graduate School; Tokushima Japan
| | - T. Nagata
- Department of Periodontology and Endodontology; Institute of Health Biosciences; Tokushima University Graduate School; Tokushima Japan
| |
Collapse
|
28
|
During A, Penel G, Hardouin P. Understanding the local actions of lipids in bone physiology. Prog Lipid Res 2015; 59:126-46. [PMID: 26118851 DOI: 10.1016/j.plipres.2015.06.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 06/12/2015] [Accepted: 06/18/2015] [Indexed: 12/19/2022]
Abstract
The adult skeleton is a metabolically active organ system that undergoes continuous remodeling to remove old and/or stressed bone (resorption) and replace it with new bone (formation) in order to maintain a constant bone mass and preserve bone strength from micro-damage accumulation. In that remodeling process, cellular balances--adipocytogenesis/osteoblastogenesis and osteoblastogenesis/osteoclastogenesis--are critical and tightly controlled by many factors, including lipids as discussed in the present review. Interest in the bone lipid area has increased as a result of in vivo evidences indicating a reciprocal relationship between bone mass and marrow adiposity. Lipids in bones are usually assumed to be present only in the bone marrow. However, the mineralized bone tissue itself also contains small amounts of lipids which might play an important role in bone physiology. Fatty acids, cholesterol, phospholipids and several endogenous metabolites (i.e., prostaglandins, oxysterols) have been purported to act on bone cell survival and functions, the bone mineralization process, and critical signaling pathways. Thus, they can be regarded as regulatory molecules important in bone health. Recently, several specific lipids derived from membrane phospholipids (i.e., sphingosine-1-phosphate, lysophosphatidic acid and different fatty acid amides) have emerged as important mediators in bone physiology and the number of such molecules will probably increase in the near future. The present paper reviews the current knowledge about: (1°) bone lipid composition in both bone marrow and mineralized tissue compartments, and (2°) local actions of lipids on bone physiology in relation to their metabolism. Understanding the roles of lipids in bone is essential to knowing how an imbalance in their signaling pathways might contribute to bone pathologies, such as osteoporosis.
Collapse
Affiliation(s)
- Alexandrine During
- Université Lille 2, Laboratoire de Physiopathologie des maladies osseuses inflammatoires (PMOI), EA4490, Faculté de Chirurgie dentaire, Lille, France.
| | - Guillaume Penel
- Université Lille 2, Laboratoire de Physiopathologie des maladies osseuses inflammatoires (PMOI), EA4490, Faculté de Chirurgie dentaire, Lille, France
| | - Pierre Hardouin
- Université Lille 2, Laboratoire de Physiopathologie des maladies osseuses inflammatoires (PMOI), EA4490, Faculté de Chirurgie dentaire, Lille, France; Université ULCO, Laboratoire de Physiopathologie des maladies osseuses inflammatoires (PMOI), EA4490, Boulogne-sur-Mer, France
| |
Collapse
|
29
|
Amato B, Compagna R, Amato M, Grande R, Butrico L, Rossi A, Naso A, Ruggiero M, de Franciscis S, Serra R. Adult vascular wall resident multipotent vascular stem cells, matrix metalloproteinases, and arterial aneurysms. Stem Cells Int 2015; 2015:434962. [PMID: 25866513 PMCID: PMC4381852 DOI: 10.1155/2015/434962] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 02/23/2015] [Accepted: 03/06/2015] [Indexed: 12/20/2022] Open
Abstract
Evidences have shown the presence of multipotent stem cells (SCs) at sites of arterial aneurysms: they can differentiate into smooth muscle cells (SMCs) and are activated after residing in a quiescent state in the vascular wall. Recent studies have implicated the role of matrix metalloproteinases in the pathogenesis of arterial aneurysms: in fact the increased synthesis of MMPs by arterial SMCs is thought to be a pivotal mechanism in aneurysm formation. The factors and signaling pathways involved in regulating wall resident SC recruitment, survival, proliferation, growth factor production, and differentiation may be also related to selective expression of different MMPs. This review explores the relationship between adult vascular wall resident multipotent vascular SCs, MMPs, and arterial aneurysms.
Collapse
Affiliation(s)
- Bruno Amato
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80100 Naples, Italy
| | - Rita Compagna
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80100 Naples, Italy
| | - Maurizio Amato
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80100 Naples, Italy
| | - Raffaele Grande
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Lucia Butrico
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Alessio Rossi
- Department of Medicine and Health Sciences, University of Molise, 88100 Campobasso, Italy
| | - Agostino Naso
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Michele Ruggiero
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Stefano de Franciscis
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
30
|
Qiu Y, Yao J, Wu X, Zhou B, Shao H, Hua T, Xiong Z, Tang G. Longitudinal assessment of oxytocin efficacy on bone and bone marrow fat masses in a rabbit osteoporosis model through 3.0-T magnetic resonance spectroscopy and micro-CT. Osteoporos Int 2015; 26:1081-92. [PMID: 25690480 DOI: 10.1007/s00198-014-2933-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 10/03/2014] [Indexed: 01/12/2023]
Abstract
UNLABELLED This study aims to longitudinally assess the effect of oxytocin on bone and bone fat masses using micro-CT, in vivo magnetic resonance spectroscopy (MRS), and histopathological adipocyte quantification. Early in vivo oxytocin (OT) treatment to the osteoporosis (OP) rabbit model may reliably inhibit bone degeneration and reduce bone marrow fat accumulation by decreasing marrow adipocyte size and density. INTRODUCTION This study aims to longitudinally assess the effect of early OT treatment on bone and bone fat masses in a rabbit OP model by comparing the results of MRS and micro-CT with histopathological findings. METHODS Sixty 20-week-old female rabbits were randomly assigned into three groups. The control and OP groups were subjected to either sham surgery or bilateral ovariectomy (OVX). The OT group was subcutaneously injected with OT daily from the second week after OVX for 8 weeks. The left proximal femurs of the rabbits were evaluated through MRS, micro-CT, and histopathological examination at 0, 4, 8, 10, and 12 weeks after operation. Differences in fat fraction (FF) values, micro-CT parameters, and calculated pathological marrow adipocytes among three groups were analyzed. RESULTS The FF values of the OP group significantly increased (p = 0.019), but the tissue mineral density (TMD) decreased (p = 0.037) from eighth week compared with those of the control group. The FF values of the OT group significantly decreased (p = 0.044), but the TMD values increased (p = 0.042) from eighth week compared with those of the OP group. The adypocyte diameter of the OT group significantly decreased (p = 0.041) from eighth week and then adypocyte density did so too from tenth week, compared with those of the OP group at the same time point. No difference in adypocyte calculation was found between the OT and control groups until the 12th week after operation. CONCLUSION Early in vivo OT treatment slowed down bone deterioration and reduced bone marrow adiposity accumulation in a rabbit OP model, which is consistent with pathologic findings. OT treatment is a promising preventive OP therapy.
Collapse
Affiliation(s)
- Y Qiu
- Department of Radiology, Shanghai Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Repercussions of NSAIDS drugs on bone tissue: the osteoblast. Life Sci 2015; 123:72-7. [PMID: 25625244 DOI: 10.1016/j.lfs.2015.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 12/30/2014] [Accepted: 01/02/2015] [Indexed: 12/11/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) can act by modulating the behavior of osteoblasts, including their proliferation, differentiation, adhesion, and migration, but not all NSAIDs have these effects. Our objective was to update the information on this issue in a review of the literature in order to offer guidance on the prescription of the appropriate NSAID(s) to patients requiring bone tissue repair. To review current knowledge of this issue by searching for all relevant publications since 2001 in the MEDLINE, EMBASE and Cochrane Library databases, we used the following descriptors: bone tissue, osteoblast, NSAIDs, Anti-inflammatory drugs. Published studies show that most NSAIDs have an adverse effect on osteoblast growth by cell cycle arrest and apoptosis induction. The effect on differentiation varies according to the drug, dose, and treatment time. Osteoblast adhesion is increased and migration decreased by some NSAIDs, such as indomethacin and diclofenac. The antigenic profile or phagocytic function can also be modulated by NSAIDs. In general, NSAIDs have an adverse effect on bone tissue and given the routine administration of NSAIDs to individuals requiring bone repair, in which the osteoblast has an essential role, this effect on bone should be borne in mind.
Collapse
|
32
|
Lee JH, Cho JY. Proteomics approaches for the studies of bone metabolism. BMB Rep 2014; 47:141-8. [PMID: 24499667 PMCID: PMC4163882 DOI: 10.5483/bmbrep.2014.47.3.270] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 12/16/2013] [Accepted: 01/04/2014] [Indexed: 01/13/2023] Open
Abstract
Bone is an active tissue, in which bone formation by osteoblast is followed by bone resorption by osteoclasts, in a repeating cycle. Proteomics approaches may allow the detection of changes in cell signal transduction, and the regulatory mechanism of cell differentiation. LC-MS/MS-based quantitative methods can be used with labeling strategies, such as SILAC, iTRAQ, TMT and enzymatic labeling. When used in combination with specific protein enrichment strategies, quantitative proteomics methods can identify various signaling molecules and modulators, and their interacting proteins in bone metabolism, to elucidate biological functions for the newly identified proteins in the cellular context. In this article, we will briefly review recent major advances in the application of proteomics for bone biology, especially from the aspect of cellular signaling. [BMB Reports 2014; 47(3): 141-148]
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Department of Veterinary Biochemistry, BK21 and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea
| | - Je-Yoel Cho
- Department of Veterinary Biochemistry, BK21 and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea
| |
Collapse
|
33
|
Pauksch L, Hartmann S, Szalay G, Alt V, Lips KS. In vitro assessment of nanosilver-functionalized PMMA bone cement on primary human mesenchymal stem cells and osteoblasts. PLoS One 2014; 9:e114740. [PMID: 25485700 PMCID: PMC4259364 DOI: 10.1371/journal.pone.0114740] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 11/13/2014] [Indexed: 01/01/2023] Open
Abstract
Peri-prosthetic infections caused by multidrug resistant bacteria have become a serious problem in surgery and orthopedics. The aim is to introduce biomaterials that avoid implant-related infections caused by multiresistant bacteria. The efficacy of silver nanoparticles (AgNP) against a broad spectrum of bacteria and against multiresistant pathogens has been repeatedly described. In the present study polymethylmethacrylate (PMMA) bone cement functionalized with AgNP and/or gentamicin were tested regarding their biocompatibility with bone forming cells. Therefore, influences on viability, cell number and differentiation of primary human mesenchymal stem cells (MSCs) and MSCs cultured in osteogenic differentiation media (MSC-OM) caused by the implant materials were studied. Furthermore, the growth behavior and the morphology of the cells on the testing material were observed. Finally, we examined the induction of cell stress, regarding antioxidative defense and endoplasmatic reticulum stress. We demonstrated similar cytocompatibility of PMMA loaded with AgNP compared to plain PMMA or PMMA loaded with gentamicin. There was no decrease in cell number, viability and osteogenic differentiation and no induction of cell stress for all three PMMA variants after 21 days. Addition of gentamicin to AgNP-loaded PMMA led to a slight decrease in osteogenic differentiation. Also an increase in cell stress was detectable for PMMA loaded with gentamicin and AgNP. In conclusion, supplementation of PMMA bone cement with gentamicin, AgNP, and both results in bone implants with an antibacterial potency and suitable cytocompatibility in MSCs and MSC-OM.
Collapse
Affiliation(s)
- Linda Pauksch
- Laboratory for Experimental Trauma Surgery, Justus-Liebig University Giessen, Giessen, Germany
| | - Sonja Hartmann
- Laboratory for Experimental Trauma Surgery, Justus-Liebig University Giessen, Giessen, Germany
| | - Gabor Szalay
- Department of Trauma Surgery, University Hospital Giessen and Marburg, Giessen, Germany
| | - Volker Alt
- Department of Trauma Surgery, University Hospital Giessen and Marburg, Giessen, Germany
| | - Katrin S. Lips
- Laboratory for Experimental Trauma Surgery, Justus-Liebig University Giessen, Giessen, Germany
- * E-mail:
| |
Collapse
|
34
|
Alves EGL, Serakides R, Boeloni JN, Rosado IR, Ocarino NM, Oliveira HP, Góes AM, Rezende CMF. Comparison of the osteogenic potential of mesenchymal stem cells from the bone marrow and adipose tissue of young dogs. BMC Vet Res 2014; 10:190. [PMID: 25178540 PMCID: PMC4236816 DOI: 10.1186/s12917-014-0190-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/13/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The aim of the present study was to compare the osteogenic potential of mesenchymal stem cells extracted from the bone marrow (BM-MSCs) and adipose tissue (AD-MSCs) of young dogs. The following parameters were assessed: dimethyl thiazolyl diphenyl tetrazolium (MTT) conversion, alkaline phosphatase (ALP) activity, collagen and mineralised matrix synthesis, and the expressions of osterix, bone sialoprotein (BSP), and osteocalcin (OC). RESULTS MTT conversion was greater in BM-MSCs compared to AD-MSCs after 14 and 21 days of differentiation; ALP activity was greater in differentiated AD-MSCs on day 7; collagen synthesis was greater in BM-MSCs on days 14 and 21; the percentage of mineralized area per field was greater in BM-MSCs compared to AD-MSCs; osterix expression was greater in BM-MSCs in days 14 and 21, and BSP and OC expression levels were greater in BM-MSCs at all the investigation time-points. CONCLUSIONS It was concluded that the osteogenic potential was greater in BM-MSCs than AD-MSCs when extracted from young dogs.
Collapse
Affiliation(s)
- Endrigo GL Alves
- Curso de Medicina Veterinária da Universidade de Uberaba (UNIUBE), Uberaba, Brazil
- Núcleo de Células Tronco e Terapia Celular Animal, NCT-TCA Departamento de Clínica e Cirurgia, Escola de veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rogéria Serakides
- Núcleo de Células Tronco e Terapia Celular Animal, NCT-TCA Departamento de Clínica e Cirurgia, Escola de veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jankerle N Boeloni
- Núcleo de Células Tronco e Terapia Celular Animal, NCT-TCA Departamento de Clínica e Cirurgia, Escola de veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabel R Rosado
- Núcleo de Células Tronco e Terapia Celular Animal, NCT-TCA Departamento de Clínica e Cirurgia, Escola de veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Natália M Ocarino
- Núcleo de Células Tronco e Terapia Celular Animal, NCT-TCA Departamento de Clínica e Cirurgia, Escola de veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Humberto P Oliveira
- Núcleo de Células Tronco e Terapia Celular Animal, NCT-TCA Departamento de Clínica e Cirurgia, Escola de veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alfredo M Góes
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cleuza MF Rezende
- Núcleo de Células Tronco e Terapia Celular Animal, NCT-TCA Departamento de Clínica e Cirurgia, Escola de veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
35
|
Oh YI, Kim JH, Kang CW. Protective effect of short-term treatment with parathyroid hormone 1-34 on oxidative stress is involved in insulin-like growth factor-I and nuclear factor erythroid 2-related factor 2 in rat bone marrow derived mesenchymal stem cells. ACTA ACUST UNITED AC 2014; 189:1-10. [PMID: 24412273 DOI: 10.1016/j.regpep.2013.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 12/24/2013] [Accepted: 12/31/2013] [Indexed: 11/30/2022]
Abstract
Bone marrow-derived mesenchymal stem cell (MSC)-mediated regeneration is a promising treatment for degenerative disease and traumatic injuries. MSCs can be isolated from rats using magnetic-activated cell sorting with CD105 antibody. We investigated the relationships between the expression of endogenous insulin-like growth factor-I (IGF-I) and nuclear factor erythroid 2-related factor 2 (Nrf-2) during short-term treatment with parathyroid hormone (PTH) 1-34-induced protective response in MSCs. PTH 1-34 (10(-9)M) decreased reactive oxygen species (ROS) generation but increased cell viability and endogenous IGF-I (p<0.01). Suppression of IGF-I and Nrf-2 using specific small interfering RNA (siRNA) blocked the effects of PTH 1-34. Furthermore, increasing cell viability of PTH against hydrogen peroxide (H2O2) was suppressed by treatment with siRNA to IGF-I and Nr-2 (p<0.05). Exogenous IGF-I (10(-9)M) also increased endogenous IGF-I, cell viability, and Nrf-2 expression. These incremental increases were lessened by Nrf-2 siRNA (p<0.05). Exogenous IGF-I also inhibited the increase of H2O2-induced ROS generation, and the decrease of PTH 1-34-induced ROS generation in the presence of IGF-I and Nrf-2 siRNA. The increase of PTH 1-34-induced Nrf-2 expression was more significant in the nucleus than in the cytosol (p<0.05). PTH 1-34 also inhibited H2O2-induced inducible nitric oxide synthase expression, but increased the expression of heme oxygenase 1/2. The results implicate PTH 1-34, Nrf-2, and IGF-I signaling pathways in the response to oxidative stress. These factors could influence IGF-I regulation of metabolic fate and survival in MSCs.
Collapse
Affiliation(s)
- Young-Il Oh
- Department of Veterinary Physiology, College of Veterinary Medicine/Bio-Safety Research Institute, Chonbuk National University, South Korea
| | - Jong-Hoon Kim
- Department of Veterinary Physiology, College of Veterinary Medicine/Bio-Safety Research Institute, Chonbuk National University, South Korea
| | - Chang-Won Kang
- Department of Veterinary Physiology, College of Veterinary Medicine/Bio-Safety Research Institute, Chonbuk National University, South Korea.
| |
Collapse
|
36
|
Yokota J, Chosa N, Sawada S, Okubo N, Takahashi N, Hasegawa T, Kondo H, Ishisaki A. PDGF-induced PI3K-mediated signaling enhances the TGF-β-induced osteogenic differentiation of human mesenchymal stem cells in a TGF-β-activated MEK-dependent manner. Int J Mol Med 2013; 33:534-42. [PMID: 24378341 PMCID: PMC3926498 DOI: 10.3892/ijmm.2013.1606] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 12/18/2013] [Indexed: 12/03/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is a critical regulator of osteogenic differentiation and the platelet-derived growth factor (PDGF) is a chemoattractant or mitogen of osteogenic mesenchymal cells. However, the combined effects of these regulators on the osteogenic differentiation of mesenchymal cells remains unknown. In this study, we investigated the effects of TGF-β and/or PDGF on the osteogenic differentiation of human mesenchymal stem cells (hMSCs). The TGF-β-induced osteogenic differentiation of UE7T-13 cells, a bone marrow-derived hMSC line, was markedly enhanced by PDGF, although PDGF alone did not induce differentiation. TGF-β induced extracellular signal-regulated kinase (ERK) phosphorylation and PDGF induced Akt phosphorylation. In addition, the mitogen-activated protein kinase (MAPK)/ERK kinase (MEK) inhibitor, U0126, suppressed the osteogenic differentiation induced by TGF-β alone. Moreover, U0126 completely suppressed the osteogenic differentiation synergistically induced by TGF-β and PDGF, whereas the phosphoinositide-3-kinase (PI3K) inhibitor, LY294002, only partially suppressed this effect. These results suggest that the enhancement of TGF-β-induced osteogenic differentiation by PDGF-induced PI3K/Akt-mediated signaling depends on TGF-β-induced MEK activity. Thus, PDGF positively modulates the TGF-β-induced osteogenic differentiation of hMSCs through synergistic crosstalk between MEK- and PI3K/Akt-mediated signaling.
Collapse
Affiliation(s)
- Jun Yokota
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028‑3694, Japan
| | - Naoyuki Chosa
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028‑3694, Japan
| | - Shunsuke Sawada
- Division of Periodontology, Department of Conservative Dentistry, Iwate Medical University School of Dentistry, Morioka, Iwate 020-8505, Japan
| | - Naoto Okubo
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028‑3694, Japan
| | - Noriko Takahashi
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028‑3694, Japan
| | - Tomokazu Hasegawa
- Department of Pediatric Dentistry, Tokushima University Hospital, Tokushima 770-8504, Japan
| | - Hisatomo Kondo
- Department of Prosthodontics and Oral Implantology, Iwate Medical University School of Dentistry, Morioka, Iwate 020-8505, Japan
| | - Akira Ishisaki
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028‑3694, Japan
| |
Collapse
|
37
|
Gigante A, Brugè F, Cecconi S, Manzotti S, Littarru GP, Tiano L. Vitamin MK-7 enhances vitamin D3-induced osteogenesis in hMSCs: modulation of key effectors in mineralization and vascularization. J Tissue Eng Regen Med 2012; 9:691-701. [DOI: 10.1002/term.1627] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 07/28/2012] [Accepted: 08/28/2012] [Indexed: 11/06/2022]
Affiliation(s)
- A. Gigante
- Dipartimento di Scienze Cliniche e Molecolari; Università Politecnica delle Marche; Ancona Italy
| | - F. Brugè
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche; Università Politecnica delle Marche; Ancona Italy
| | - S. Cecconi
- Dipartimento di Scienze Cliniche e Molecolari; Università Politecnica delle Marche; Ancona Italy
| | - S. Manzotti
- Dipartimento di Scienze Cliniche e Molecolari; Università Politecnica delle Marche; Ancona Italy
| | - G. P. Littarru
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche; Università Politecnica delle Marche; Ancona Italy
| | - L. Tiano
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche; Università Politecnica delle Marche; Ancona Italy
| |
Collapse
|
38
|
Kim JM, Kim J, Kim YH, Kim KT, Ryu SH, Lee TG, Suh PG. Comparative secretome analysis of human bone marrow-derived mesenchymal stem cells during osteogenesis. J Cell Physiol 2012; 228:216-24. [DOI: 10.1002/jcp.24123] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
39
|
Giovanini AF, Grossi JRA, Gonzaga CC, Zielak JC, Göhringer I, Vieira JDS, Kuczera J, de Oliveira Filho MA, Deliberador TM. Leukocyte-Platelet-Rich Plasma (L-PRP) Induces an Abnormal Histophenotype in Craniofacial Bone Repair Associated with Changes in the Immunopositivity of the Hematopoietic Clusters of Differentiation, Osteoproteins, and TGF-β1. Clin Implant Dent Relat Res 2012; 16:259-72. [DOI: 10.1111/j.1708-8208.2012.00478.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
| | | | | | - João Cesar Zielak
- Masters Program in Clinical Dentistry; Positivo University; Curitiba Brazil
| | - Isabella Göhringer
- Masters Program in Clinical Dentistry; Positivo University; Curitiba Brazil
| | | | | | | | | |
Collapse
|
40
|
Gioia R, Panaroni C, Besio R, Palladini G, Merlini G, Giansanti V, Scovassi IA, Villani S, Villa I, Villa A, Vezzoni P, Tenni R, Rossi A, Marini JC, Forlino A. Impaired osteoblastogenesis in a murine model of dominant osteogenesis imperfecta: a new target for osteogenesis imperfecta pharmacological therapy. Stem Cells 2012; 30:1465-76. [PMID: 22511244 PMCID: PMC3459187 DOI: 10.1002/stem.1107] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The molecular basis underlying the clinical phenotype in bone diseases is customarily associated with abnormal extracellular matrix structure and/or properties. More recently, cellular malfunction has been identified as a concomitant causative factor and increased attention has focused on stem cells differentiation. Classic osteogenesis imperfecta (OI) is a prototype for heritable bone dysplasias: it has dominant genetic transmission and is caused by mutations in the genes coding for collagen I, the most abundant protein in bone. Using the Brtl mouse, a well-characterized knockin model for moderately severe dominant OI, we demonstrated an impairment in the differentiation of bone marrow progenitor cells toward osteoblasts. In mutant mesenchymal stem cells (MSCs), the expression of early (Runx2 and Sp7) and late (Col1a1 and Ibsp) osteoblastic markers was significantly reduced with respect to wild type (WT). Conversely, mutant MSCs generated more colony-forming unit-adipocytes compared to WT, with more adipocytes per colony, and increased number and size of triglyceride drops per cell. Autophagy upregulation was also demonstrated in mutant adult MSCs differentiating toward osteogenic lineage as consequence of endoplasmic reticulum stress due to mutant collagen retention. Treatment of the Brtl mice with the proteasome inhibitor Bortezomib ameliorated both osteoblast differentiation in vitro and bone properties in vivo as demonstrated by colony-forming unit-osteoblasts assay and peripheral quantitative computed tomography analysis on long bones, respectively. This is the first report of impaired MSC differentiation to osteoblasts in OI, and it identifies a new potential target for the pharmacological treatment of the disorder.
Collapse
Affiliation(s)
- Roberta Gioia
- Department of Molecular Medicine, Section of Biochemistry, University of Pavia, Pavia, Italy
| | - Cristina Panaroni
- Department of Molecular Medicine, Section of Biochemistry, University of Pavia, Pavia, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, CNR, Milan, Italy
| | - Roberta Besio
- Department of Molecular Medicine, Section of Biochemistry, University of Pavia, Pavia, Italy
| | - Giovanni Palladini
- Department of Molecular Medicine, Section of Biochemistry, University of Pavia, Pavia, Italy
- Amyloidosis Research and Treatment Center, Biotechnology Research Laboratories, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giampaolo Merlini
- Department of Molecular Medicine, Section of Biochemistry, University of Pavia, Pavia, Italy
- Amyloidosis Research and Treatment Center, Biotechnology Research Laboratories, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | | | - Simona Villani
- Department of Health Sciences, Section of Medical Statistic and Epidemiology, University of Pavia, Pavia, Italy
| | - Isabella Villa
- Bone Metabolic Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Anna Villa
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, CNR, Milan, Italy
- Istituto Clinico Humanitas, Rozzano, Italy
| | - Paolo Vezzoni
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, CNR, Milan, Italy
- Istituto Clinico Humanitas, Rozzano, Italy
| | - Ruggero Tenni
- Department of Molecular Medicine, Section of Biochemistry, University of Pavia, Pavia, Italy
| | - Antonio Rossi
- Department of Molecular Medicine, Section of Biochemistry, University of Pavia, Pavia, Italy
| | - Joan C. Marini
- Bone and Extracellular Matrix Branch, NICHD, NIH, Bethesda, Maryland, USA
| | - Antonella Forlino
- Department of Molecular Medicine, Section of Biochemistry, University of Pavia, Pavia, Italy
| |
Collapse
|
41
|
Jiang L, Lu X, Leng Y, Qu S, Feng B, Weng J, Watari F. Osteoblast behavior on TiO2 microgrooves prepared by soft-lithography and sol–gel methods. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2012. [DOI: 10.1016/j.msec.2012.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
42
|
Gimble JM, Nuttall ME. The relationship between adipose tissue and bone metabolism. Clin Biochem 2012; 45:874-9. [PMID: 22429519 DOI: 10.1016/j.clinbiochem.2012.03.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 02/28/2012] [Accepted: 03/01/2012] [Indexed: 01/06/2023]
Abstract
OBJECTIVES The authors have set out to evaluate the literature relevant to the dynamic regulation of adipogenesis and osteogenesis. DESIGN AND METHODS A detailed search of the past and recent literature was conducted on Pubmed using a combination of keywords including: adipogenesis, bone marrow, hematopoiesis, mesenchymal stromal/stem cell, and osteogenesis. RESULTS Throughout one's lifespan, the bone marrow microenvironment provides a unique niche for mesenchymal stromal/stem cells (BMSCs) and hematopoietic stem cells (HSCs). The marrow changes as a function of biological age and pathophysiology. Historically, clinical biochemistry has observed these changes from an HSC and hematological perspective. Nevertheless, these changes also reflect the balance between BMSC adipogenic and osteogenic processes which can display an inverse or reciprocal relationship. Multiple hormonal factors and nuclear hormone receptor ligands and drugs are responsible for BMSC lineage selection. Data from a number of laboratories now implicates endocrine feedback loops between extramedullary adipose depots and the central nervous system. CONCLUSIONS This concise review provides a perspective on the mechanisms regulating BMSC differentiation in the context of biological aging, obesity, and osteoporosis.
Collapse
Affiliation(s)
- Jeffrey M Gimble
- Stem Cell Biology Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA.
| | | |
Collapse
|
43
|
Schoengraf P, Lambris JD, Recknagel S, Kreja L, Liedert A, Brenner RE, Huber-Lang M, Ignatius A. Does complement play a role in bone development and regeneration? Immunobiology 2012; 218:1-9. [PMID: 22464814 DOI: 10.1016/j.imbio.2012.01.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 01/27/2012] [Accepted: 01/27/2012] [Indexed: 12/16/2022]
Abstract
The skeletal and the immune system are not two independent systems, rather, there are multifaceted and complex interactions between the different cell types of both systems and there are several shared cytokines. As a part of the innate immunity, the complement system was found to be an important link between bone and immunity. Complement proteins appear to be involved in bone development and homeostasis, and specifically influence osteoblast and osteoclast activity. This review describes the complex mutual regulation of the two systems, and indicates some of the negative side effects as a result of inappropriate or excessive complement activation.
Collapse
Affiliation(s)
- Philipp Schoengraf
- Institute of Orthopaedic Research and Biomechanics, Centre of Muskuloskelettal Research, University of Ulm, Helmholtzstrasse 14, 89081 Ulm, Germany
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Illich DJ, Demir N, Stojković M, Scheer M, Rothamel D, Neugebauer J, Hescheler J, Zöller JE. Concise review: induced pluripotent stem cells and lineage reprogramming: prospects for bone regeneration. Stem Cells 2011; 29:555-63. [PMID: 21308867 DOI: 10.1002/stem.611] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bone tissue for transplantation therapies is in high demand in clinics. Osteodegenerative diseases, in particular, osteoporosis and osteoarthritis, represent serious public health issues affecting a respectable proportion of the elderly population. Furthermore, congenital indispositions from the spectrum of craniofacial malformations such as cleft palates and systemic disorders including osteogenesis imperfecta are further increasing the need for bone tissue. Additionally, the reconstruction of fractured bone elements after accidents and the consumption of bone parts during surgical tumor excisions represent frequent clinical situations with deficient availability of healthy bone tissue for therapeutic transplantations. Epigenetic reprogramming represents a powerful technology for the generation of healthy patient-specific cells to replace or repair diseased or damaged tissue. The recent generation of induced pluripotent stem cells (iPSCs) is probably the most promising among these approaches dominating the literature of current stem cell research. It allows the generation of pluripotent stem cells from adult human skin cells from which potentially all cell types of the human body could be obtained. Another technique to produce clinically interesting cell types is direct lineage reprogramming (LR) with the additional advantage that it can be applied directly in vivo to reconstitute a damaged organ. Here, we want to present the two technologies of iPSCs and LR, to outline the current states of research, and to discuss possible strategies for their implementation in bone regeneration.
Collapse
Affiliation(s)
- Damir J Illich
- Medical Center, Institute for Neurophysiology, University of Cologne, Cologne, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Jones E, Yang X. Mesenchymal stem cells and bone regeneration: current status. Injury 2011; 42:562-8. [PMID: 21489533 DOI: 10.1016/j.injury.2011.03.030] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 03/17/2011] [Indexed: 02/02/2023]
Abstract
The enhancement of bone regeneration with biological agents including osteogenic growth factors and mesenchymal stem cells (MSCs) is becoming a clinical reality. Many exciting findings have been obtained following MSC implantation in animal models, and the data demonstrating their clinical efficacy in humans are promising. The overwhelming majority of experimental work has been performed with MSCs "amplified"in vitro. The nature of native MSCs in skeletal tissues however, remains poorly understood. This review summarizes recent findings pertaining to the definition and characterisation of MSCs in skeletal tissues and discusses the mechanisms of their actions in regenerating of bone in vivo. In respect to traditional tissue engineering paradigm, we bring together literature showing that the ways MSCs are extracted, expanded and implanted can considerably affect bone formation outcomes. Additionally, we discuss current animal models used in MSC research and highlight recent experiments showing important contribution of the host, and not only donor MSCs, in bone tissue formation. This knowledge provides a platform for novel therapy development for bone regeneration based on pharmacologically manipulated endogenous MSCs.
Collapse
Affiliation(s)
- Elena Jones
- Rheumatology, Mesenchymal Stem Cell Biology Group, Academic Unit of Musculoskeletal Disease, Leeds Institute of Molecular Medicine, St James's University Hospital, University of Leeds, Beckett Street, Leeds LS9 7TF, United Kingdom.
| | | |
Collapse
|
46
|
Yu H, de Vos P, Ren Y. Overexpression of osteoprotegerin promotes preosteoblast differentiation to mature osteoblasts. Angle Orthod 2011; 81:100-106. [PMID: 20936961 DOI: 10.2319/050210-238.1] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE The hypothesis of the present study is that overexpression of osteoprotegerin (OPG) promotes preosteoblast maturation. MATERIALS AND METHODS The preosteoblast cell line MC3T3-E1 was transfected with OPG overexpression. OPG expression was confirmed by enzyme-linked immunosorbent assay (ELISA) and Western blot. Changes in the transcription factors in OPG-expressing cells were assessed by real-time polymerase quantitative polymerase chain reaction (RT-qPCR). Alkaline phosphate (ALP) expression was measured by ELISA. RESULTS The success of stable transfection of MC3T3-E1 cells with OPG overexpression was confirmed by MoFlow sorting followed by G418 selection. RT-qPCR showed that expression of RunX2, the most important osteoblast differentiation controlling factor, was suppressed. Smad1 and Akt1, as well as ALP, were upregulated in the OPG overexpressing cells. CONCLUSION Results from the present study provide evidence that overexpression of OPG in preosteoblasts promotes its differentiation into mature osteoblasts.
Collapse
Affiliation(s)
- Hongyou Yu
- Department of Orthodontics, University Medical Centre Groningen, University of Groningen, The Netherlands
| | | | | |
Collapse
|
47
|
Syed FA, Hoey KA. Integrative physiology of the aging bone: insights from animal and cellular models. Ann N Y Acad Sci 2010; 1211:95-106. [PMID: 21062298 DOI: 10.1111/j.1749-6632.2010.05813.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Age-related bone loss is a common worldwide phenomenon in the aging population, placing them at an increased risk of fractures. Fortunately, basic and translational studies have been pivotal in providing us with a mechanistic understanding of the cellular and molecular pathophysiology of this condition. This review focuses on the current concepts and paradigms of age-related bone loss and how various animal and cellular models have broadened our understanding in this fascinating but complex area. Changes in hormonal, neuronal, and biochemical cues with age and their effect on bone have been discussed. This review also outlines recent studies on the relationship between bone and fat in the marrow, as well as the fate of the marrow mesenchymal stromal cell population, which can give rise to either bone-forming osteoblasts or fat-forming adipocytic cells as a function of age.
Collapse
Affiliation(s)
- Farhan A Syed
- Abbott Bioresearch Center, Worcester, Massachusetts, USA.
| | | |
Collapse
|
48
|
Giovanini AF, Deliberador TM, Gonzaga CC, de Oliveira Filho MA, Göhringer I, Kuczera J, Zielak JC, de Andrade Urban C. Platelet-rich plasma diminishes calvarial bone repair associated with alterations in collagen matrix composition and elevated CD34+ cell prevalence. Bone 2010; 46:1597-603. [PMID: 20206725 DOI: 10.1016/j.bone.2010.02.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 02/11/2010] [Accepted: 02/25/2010] [Indexed: 12/12/2022]
Abstract
The interaction between platelets and both type I and III collagens plays an important role in modulating platelet adhesion and aggregation, also contributing to the chemotaxis of CD34+ cells. The interaction with type III collagen can maintain high levels of collagen and alter the biology of bone repair when the PRP is used. The aim of this study was to evaluate the effect of platelet-rich plasma (PRP) and autograft on the presence of type III and type I collagens, the ratio between them, as well as the presence of CD34+ progenitor cells, while comparing these results by means of a histomorphometric analysis of the bone tissue. Four bone defects (8.0mm in diameter and 2.0mm in depth) were produced on the calvarium of 23 rabbits. The surgical defects were treated with either autogenous bone grafts, autogenous bone grafts with PRP and PRP alone. Animals were euthanized at 2, 4 or 6 weeks post-surgery. Histological, histomorphometric and immunohistochemical analyses were performed to assess repair time, as well as the expression of type I and III collagens, and number of progenitor CD34+ cells. Data were analyzed using the ANOVA and Student-Newman-Keuls test (alpha=5%). An enlarged granulation and medullary tissue areas in the PRP groups were observed. The use of PRP in this study hindered bone deposition, also enhanced type III to type I collagen ratio and the chemotaxis of CD34+ progenitor cells, similarly to a thrombogenic effect.
Collapse
|
49
|
Luvizuto ER, Dias SMD, Queiroz TP, Okamoto T, Garcia IR, Okamoto R, Dornelles RCM. Osteocalcin immunolabeling during the alveolar healing process in ovariectomized rats treated with estrogen or raloxifene. Bone 2010; 46:1021-9. [PMID: 20036346 DOI: 10.1016/j.bone.2009.12.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 12/03/2009] [Accepted: 12/16/2009] [Indexed: 10/20/2022]
Abstract
The influence of an estrogen-deficient state was evaluated in this study and also its treatments with estrogen (E(2)) or with raloxifene (RLX) on the expression of osteocalcin during the periods of the chronology of the alveolar bone healing process (7, 14, 21, 28 and 42 post-extraction days) by means of immunohistochemistry reactions and histomorphometric analysis. Rats (200-220 g) with oestrus cycles normal were either OVX or sham-operated and divided into four groups: sham, OVX control (OVX/O), estrogen (OVX/E(2); 17 beta-estradiol, 400 microg/mo) and raloxifene (OVX/RLX; 1 mg/kg bw/d) groups. Histomorphometric analysis showed the sham group presented the highest mean value of bone formation post-extraction. The reaction of immunohistochemistry for osteocalcin presented stronger expression of osteocalcin with predominance at 14 and 21 days on sham group. The OVX/RLX group presented better results than OVX/E(2), considering the expression of osteocalcin in osteoblastic lineage cells, but still inferior than the sham group. It was concluded that ovariectomy decreases the mineralization process and the osteocalcin expression during the chronology of the alveolar healing process that is not totally recovered with estrogen replacement or raloxifene treatment.
Collapse
|
50
|
Lindner U, Kramer J, Rohwedel J, Schlenke P. Mesenchymal Stem or Stromal Cells: Toward a Better Understanding of Their Biology? ACTA ACUST UNITED AC 2010; 37:75-83. [PMID: 20737049 DOI: 10.1159/000290897] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 02/24/2010] [Indexed: 01/12/2023]
Abstract
The adult bone marrow has been generally considered to be composed of hematopoietic tissue and the associated supporting stroma. Within the latter compartment, a subset of cells with multipotent differentiation capacity exists, usually referred to as mesenchymal stem cells. Mesenchymal stem cells can easily be expanded ex vivo and induced to differentiate into several cell types, including osteoblasts, adipocytes and chondrocytes. Up to now, mesenchymal stem cells have gained wide popularity. Despite the rapid growth in this field, irritations remain with respect to the defining characteristics of these cells, including their differentiation potency, self-renewal and in vivo properties. As a consequence, there is a growing tendency to challenge the term mesenchymal stem cell, especially with respect to the stem cell characteristics. Here, we revisit the experimental origins of mesenchymal stem cells, their classical differentiation capacity into mesodermal lineages and their immunophenotype in order to assess their stemness and function. Based on these essentials, it has to be revisited if the designation as a stem cell remains an appropriate term.
Collapse
Affiliation(s)
- Ulrich Lindner
- Medical Department I, Division of Nephrology and Transplantation Unit, University of Lübeck, Germany
| | | | | | | |
Collapse
|