51
|
Yang F, Jia Y, Sun Q, Zheng C, Liu C, Wang W, Du L, Kang S, Niu X, Li J. Raloxifene improves TNF-α-induced osteogenic differentiation inhibition of bone marrow mesenchymal stem cells and alleviates osteoporosis. Exp Ther Med 2020; 20:309-314. [PMID: 32550885 PMCID: PMC7296296 DOI: 10.3892/etm.2020.8689] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/09/2019] [Indexed: 01/01/2023] Open
Abstract
Effect of raloxifene (RLF) on the improvement of inhibited osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) resulted from tumor necrosis factor-α (TNF-α) induction, thus alleviating the progression of osteoporosis (OP), was investigated. An in vivo OP rat model was constructed by performing the procedures of ovariectomy (OVX). Rats were randomly divided into sham group, OVX group and RLF+OVX group. BMSCs were extracted from healthy rats, and randomly divided into control group, TNF-α group, RLF group and TNF-α+RLF group. Viability and cellular calcification ability in each group were detected. The relative levels of osteocalcin (OCN), Runx2 and NF-κB in cells with different treatments were determined. The body weight of rats in the OVX group and RLF+OVX group gradually increased compared with that in the sham group on the 8th week. No significant difference in body weight was observed between the rats of the OVX group and RLF+OVX group. Bone metabolism index (BMD) in the rats of the RLF+OVX group was higher than that of the OVX group, and lower compared with that of the sham group. Compared with the sham group, the elastic/max radial degree and elastic/max load of femora were reduced in the OVX group and RLF+OVX group, especially in the OVX group. The relative levels of OCN and Runx2, as well as the ALP activity and calcification ability, were decreased in the OVX group compared with the sham group, and the effect was partially reversed by the RLF treatment. After osteogenic differentiation of BMSCs, the viability and calcification ability were markedly reduced in TNF-α group, which was reversed by RLF treatment. Moreover, TNF-α induction downregulated the relative levels of OCN and Runx2, and RLF treatment could enhance their levels. The upregulated NF-κB protein level, induced by TNF-α, was reduced after RLF treatment. TNF-α induction inhibits osteogenic differentiation of BMSCs, which could be remarkably alleviated by RLF. It is suggested that RLF contributes to the alleviation of OP progression.
Collapse
Affiliation(s)
- Fenghe Yang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P.R. China.,Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing 100083, P.R. China
| | - Yusong Jia
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Qi Sun
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Chenying Zheng
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Chuyin Liu
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Wei Wang
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Li Du
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Shengqian Kang
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Xufeng Niu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P.R. China.,Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing 100083, P.R. China
| | - Jinyu Li
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| |
Collapse
|
52
|
Pathophysiological and Genetic Aspects of Vascular Calcification. Cardiol Res Pract 2020; 2020:5169069. [PMID: 32411445 PMCID: PMC7201852 DOI: 10.1155/2020/5169069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/17/2020] [Accepted: 03/23/2020] [Indexed: 12/21/2022] Open
Abstract
Recent evidence suggests that vascular calcification is an independent cardiovascular risk factor (CRF) of morbidity and mortality. New studies point out the existence of a complex physiopathological mechanism that involves inflammation, oxidation, the release of chemical mediators, and genetic factors that promote the osteochondrogenic differentiation of vascular smooth muscle cells (VSMC). This review will evaluate the main mechanisms involved in the pathophysiology and genetics modulation of the process of vascular calcification. Objective. A systematic review of the pathophysiology factors involved in vascular calcification and its genetic influence was performed. Methods. A systematic review was conducted in the Medline and PubMed databases and were searched for studies concerning vascular calcification using the keywords and studies published until 2020/01 in English. Inclusion Criteria. Studies in vitro, animal models, and humans. These include cohort (both retrospective and prospective cohort studies), case-control, cross-sectional, and systematic reviews. Exclusion Criteria. Studies before 2003 of the existing literature.
Collapse
|
53
|
Bessueille L, Briolay A, Como J, Mebarek S, Mansouri C, Gleizes M, El Jamal A, Buchet R, Dumontet C, Matera EL, Mornet E, Millan JL, Fonta C, Magne D. Tissue-nonspecific alkaline phosphatase is an anti-inflammatory nucleotidase. Bone 2020; 133:115262. [PMID: 32028019 PMCID: PMC7185042 DOI: 10.1016/j.bone.2020.115262] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/16/2020] [Accepted: 02/03/2020] [Indexed: 12/11/2022]
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is necessary for skeletal mineralization by its ability to hydrolyze the mineralization inhibitor inorganic pyrophosphate (PPi), which is mainly generated from extracellular ATP by ectonucleotide pyrophosphatase phosphodiesterase 1 (NPP1). Since children with TNAP deficiency develop bone metaphyseal auto-inflammations in addition to rickets, we hypothesized that TNAP also exerts anti-inflammatory effects relying on the hydrolysis of pro-inflammatory adenosine nucleotides into the anti-inflammatory adenosine. We explored this hypothesis in bone metaphyses of 7-day-old Alpl+/- mice (encoding TNAP), in mineralizing hypertrophic chondrocytes and osteoblasts, and non-mineralizing mesenchymal stem cells (MSCs) and neutrophils, which express TNAP and are present, or can be recruited in the metaphysis. Bone metaphyses of 7-day-old Alpl+/- mice had significantly increased levels of Il-1β and Il-6 and decreased levels of the anti-inflammatory Il-10 cytokine as compared with Alpl+/+ mice. In bone metaphyses, murine hypertrophic chondrocytes and osteoblasts, Alpl mRNA levels were much higher than those of the adenosine nucleotidases Npp1, Cd39 and Cd73. In hypertrophic chondrocytes, inhibition of TNAP with 25 μM of MLS-0038949 decreased the hydrolysis of AMP and ATP. However, TNAP inhibition did not significantly modulate ATP- and adenosine-associated effects in these cells. We observed that part of TNAP proteins in hypertrophic chondrocytes was sent from the cell membrane to matrix vesicles, which may explain why TNAP participated in the hydrolysis of ATP but did not significantly modulate its autocrine pro-inflammatory effects. In MSCs, TNAP did not participate in ATP hydrolysis nor in secretion of inflammatory mediators. In contrast, in neutrophils, TNAP inhibition with MLS-0038949 significantly exacerbated ATP-associated activation and secretion of IL-1β, and extended cell survival. Collectively, these results demonstrate that TNAP is a nucleotidase in both hypertrophic chondrocytes and neutrophils, and that this nucleotidase function is associated with autocrine effects on inflammation only in neutrophils.
Collapse
Affiliation(s)
- L Bessueille
- Univ Lyon; University Lyon 1; ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - A Briolay
- Univ Lyon; University Lyon 1; ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - J Como
- Univ Lyon; University Lyon 1; ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - S Mebarek
- Univ Lyon; University Lyon 1; ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - C Mansouri
- Univ Lyon; University Lyon 1; ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - M Gleizes
- Centre de recherche cerveau et cognition (CERCO), UMR CNRS 5549 université de Toulouse, UPS, France
| | - A El Jamal
- Univ Lyon; University Lyon 1; ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - R Buchet
- Univ Lyon; University Lyon 1; ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - C Dumontet
- Anticancer Antibodies, CRCL, INSERM U1052, CNRS UMR 5286, CLB, UCBL, Lyon, France
| | - E L Matera
- Anticancer Antibodies, CRCL, INSERM U1052, CNRS UMR 5286, CLB, UCBL, Lyon, France
| | - E Mornet
- Service de biologie, unité de génétique constitutionnelle, centre hospitalier de Versailles, Le Chesnay, France
| | - J L Millan
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - C Fonta
- Centre de recherche cerveau et cognition (CERCO), UMR CNRS 5549 université de Toulouse, UPS, France
| | - D Magne
- Univ Lyon; University Lyon 1; ICBMS, UMR CNRS 5246, F-69622 Lyon, France.
| |
Collapse
|
54
|
Shimo T, Takebe H, Okui T, Kunisada Y, Ibaragi S, Obata K, Kurio N, Shamsoon K, Fujii S, Hosoya A, Irie K, Sasaki A, Iwamoto M. Expression and Role of IL-1β Signaling in Chondrocytes Associated with Retinoid Signaling during Fracture Healing. Int J Mol Sci 2020; 21:ijms21072365. [PMID: 32235405 PMCID: PMC7177407 DOI: 10.3390/ijms21072365] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/15/2020] [Accepted: 03/26/2020] [Indexed: 01/08/2023] Open
Abstract
The process of fracture healing consists of an inflammatory reaction and cartilage and bone tissue reconstruction. The inflammatory cytokine interleukin-1β (IL-1β) signal is an important major factor in fracture healing, whereas its relevance to retinoid receptor (an RAR inverse agonist, which promotes endochondral bone formation) remains unclear. Herein, we investigated the expressions of IL-1β and retinoic acid receptor gamma (RARγ) in a rat fracture model and the effects of IL-1β in the presence of one of several RAR inverse agonists on chondrocytes. An immunohistochemical analysis revealed that IL-1β and RARγ were expressed in chondrocytes at the fracture site in the rat ribs on day 7 post-fracture. In chondrogenic ATDC5 cells, IL-1β decreases the levels of aggrecan and type II collagen but significantly increased the metalloproteinase-13 (Mmp13) mRNA by real-time reverse transcription-polymerase chain reaction (RT-PCR) analysis. An RAR inverse agonist (AGN194310) inhibited IL-1β-stimulated Mmp13 and Ccn2 mRNA in a dose-dependent manner. Phosphorylated extracellular signal regulated-kinases (pERK1/2) and p-p38 mitogen-activated protein kinase (MAPK) were increased time-dependently by IL-1β treatment, and the IL-1β-induced p-p38 MAPK was inhibited by AGN194310. Experimental p38 inhibition led to a drop in the IL-1β-stimulated expressions of Mmp13 and Ccn2 mRNA. MMP13, CCN2, and p-p38 MAPK were expressed in hypertrophic chondrocytes near the invaded vascular endothelial cells. As a whole, these results point to role of the IL-1β via p38 MAPK as important signaling in the regulation of the endochondral bone formation in fracture healing, and to the actions of RAR inverse agonists as potentially relevant modulators of this process.
Collapse
Affiliation(s)
- Tsuyoshi Shimo
- Division of Reconstructive Surgery for Oral and Maxillofacial Region, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido 061-0293, Japan;
- Correspondence: ; Tel./Fax: +81-133-23-1429
| | - Hiroaki Takebe
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido 061-0293, Japan; (H.T.); (A.H.); (K.I.)
| | - Tatsuo Okui
- Departments of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8525, Japan; (T.O.); (Y.K.); (S.I.); (K.O.); (A.S.)
| | - Yuki Kunisada
- Departments of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8525, Japan; (T.O.); (Y.K.); (S.I.); (K.O.); (A.S.)
| | - Soichiro Ibaragi
- Departments of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8525, Japan; (T.O.); (Y.K.); (S.I.); (K.O.); (A.S.)
| | - Kyoichi Obata
- Departments of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8525, Japan; (T.O.); (Y.K.); (S.I.); (K.O.); (A.S.)
| | - Naito Kurio
- Department of Oral Surgery, Tokushima University Graduate School, Tokushima 770-8504, Japan;
| | - Karnoon Shamsoon
- Division of Clinical Cariology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, University of Hokkaido, School of Dentistry, Hokkaido 061-0293, Japan;
| | - Saki Fujii
- Division of Reconstructive Surgery for Oral and Maxillofacial Region, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido 061-0293, Japan;
| | - Akihiro Hosoya
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido 061-0293, Japan; (H.T.); (A.H.); (K.I.)
| | - Kazuharu Irie
- Division of Histology, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido 061-0293, Japan; (H.T.); (A.H.); (K.I.)
| | - Akira Sasaki
- Departments of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8525, Japan; (T.O.); (Y.K.); (S.I.); (K.O.); (A.S.)
| | - Masahiro Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| |
Collapse
|
55
|
Chen B, You Y, Ma A, Song Y, Jiao J, Song L, Shi E, Zhong X, Li Y, Li C. Zn-Incorporated TiO 2 Nanotube Surface Improves Osteogenesis Ability Through Influencing Immunomodulatory Function of Macrophages. Int J Nanomedicine 2020; 15:2095-2118. [PMID: 32273705 PMCID: PMC7109325 DOI: 10.2147/ijn.s244349] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 02/29/2020] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Zinc (Zn), an essential trace element in the body, has stable chemical properties, excellent osteogenic ability and moderate immunomodulatory property. In the present study, a Zn-incorporated TiO2 nanotube (TNT) was fabricated on titanium (Ti) implant material. We aimed to evaluate the influence of nano-scale topography and Zn on behaviors of murine RAW 264.7 macrophages. Moreover, the effects of Zn-incorporated TNT surface-regulated macrophages on the behaviors and osteogenic differentiation of murine MC3T3-E1 osteoblasts were also investigated. METHODS TNT coatings were firstly fabricated on a pure Ti surface using anodic oxidation, and then nano-scale Zn particles were incorporated onto TNTs by the hydrothermal method. Surface topography, chemical composition, roughness, hydrophilicity, Zn release pattern and protein adsorption ability of the Zn-incorporated TiO2 nanotube surface were characterized by scanning electron microscopy (SEM), X-ray diffraction (XRD) and X-ray photoelectron spectroscopy (XPS), surface profiler, contact angle test, Zn release test and protein adsorption test. The cell behaviors and both pro-inflammatory (M1) and pro-regenerative (M2) marker gene and protein levels in macrophages cultured on Zn-incorporated TNTs surfaces with different TNT diameters were detected. The supernatants of macrophages were extracted and preserved as conditioned medium (CM). Furthermore, the behaviors and osteogenic properties of osteoblasts cultured in CM on various surfaces were evaluated. RESULTS The release profile of Zn on Zn-incorporated TNT surfaces revealed a controlled release pattern. Macrophages cultured on Zn-incorporated TNT surfaces displayed enhanced gene and protein expression of M2 markers, and M1 markers were moderately inhibited, compared with the LPS group (the inflammation model). When cultured in CM, osteoblasts cultured on Zn-incorporated TNTs showed strengthened cell proliferation, adhesion, osteogenesis-related gene expression, alkaline phosphatase activity and extracellular mineralization, compared with their TNT counterparts and the Ti group. CONCLUSION This study suggests that the application of Zn-incorporated TNT surfaces may establish an osteogenic microenvironment and accelerate bone formation. It provided a promising strategy of Ti surface modification for a better applicable prospect.
Collapse
Affiliation(s)
- Bo Chen
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yapeng You
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Aobo Ma
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yunjia Song
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Jian Jiao
- Department of Stomatology, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Liting Song
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Enyu Shi
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Xue Zhong
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Ying Li
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Changyi Li
- School of Dentistry, Stomatological Hospital, Tianjin Medical University, Tianjin, People’s Republic of China
| |
Collapse
|
56
|
Jia Y, He W, Zhang H, He L, Wang Y, Zhang T, Peng J, Sun P, Qian Y. Morusin Ameliorates IL-1β-Induced Chondrocyte Inflammation and Osteoarthritis via NF-κB Signal Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1227-1240. [PMID: 32273685 PMCID: PMC7105369 DOI: 10.2147/dddt.s244462] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
Purpose Osteoarthritis (OA) is one of the most common degenerative joint diseases in the world, characterized primarily by the progressive degradation of articular cartilage. Accumulating evidence has shown that Morusin, a flavonoid derived from the root bark of Morus alba (mulberry) plants, exerts unique protective properties in several diseases. However, its effects on OA, specifically, have not yet been characterized. Methods In this study, we evaluated the anti-inflammatory effect of Morusin on mouse chondrocytes and its underlying mechanism in vitro. In addition, the protective effect of Morusin on destabilization of the medial meniscus (DMM) model was also explored in vivo. Results In vitro, IL-1β-induced activation of inflammatory factors (TNF-α, IL-6, INOS and COX2) was dramatically suppressed by Morusin. Further, Morusin treatment inhibited the expression of ADAMTS5 and metalloproteinase (MMPs), both of which regulate extracellular matrix degradation. Morusin also decreased IL-1β-induced p65 phosphorylation and IκBα degradation. In vivo, degradation of the articular cartilage following surgical DMM, which mimicked OA pathology, was abrogated following treatment with Morusin, thus demonstrating a protective effect in the DMM model. Conclusion Herein, we demonstrate that Morusin reduces the OA inflammatory response in vitro and protects against articular cartilage degradation in vivo potentially via regulation of the NF-κB pathway. Hence, Morusin may prove to be an effective candidate for novel OA therapeutic strategies.
Collapse
Affiliation(s)
- Yewei Jia
- Department of Orthopaedics, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang 312000, People's Republic of China
| | - Wei He
- Department of Orthopaedics, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang 312000, People's Republic of China
| | - Hanxiao Zhang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Lei He
- Department of Orthopaedics, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang 312000, People's Republic of China
| | - Yanben Wang
- Department of Orthopaedics, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang 312000, People's Republic of China
| | - Tan Zhang
- Department of Orthopaedics, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang 312000, People's Republic of China
| | - Jiaxuan Peng
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Guangxi 530021, People's Republic of China
| | - Peng Sun
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Yu Qian
- Department of Orthopaedics, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang 312000, People's Republic of China
| |
Collapse
|
57
|
Wang J, Chen X, Yang X, Guo B, Li D, Zhu X, Zhang X. Positive role of calcium phosphate ceramics regulated inflammation in the osteogenic differentiation of mesenchymal stem cells. J Biomed Mater Res A 2020; 108:1305-1320. [PMID: 32064734 DOI: 10.1002/jbm.a.36903] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/09/2020] [Accepted: 02/12/2020] [Indexed: 02/05/2023]
Abstract
Recently, researches have confirmed the crucial role of inflammatory response in Ca-P ceramic-induced osteogenesis, however, the underlying mechanism has not yet been fully understood. In this study, BCP and β-TCP ceramics were used as material models to investigate the effect of physicochemical properties on inflammatory response in vitro. The results showed that BCP and β-TCP could support macrophages attachment, proliferation, and spreading favorably, as well as promote gene expressions of inflammatory related cytokines (IL-1, IL-6, MCP-1, and TNF-α) and growth factors (TGF-β, FGF, PDGF, VEGF, IGF, and EGF). BCP showed a facilitating function on the gene expressions earlier than β-TCP. Further coculture experiments performed in vitro demonstrated that the CMs containing various increased cytokines for macrophages pre-culture could significantly promote MSCs osteogenic differentiation, which was confirmed by the gene expressions of osteogenic specific markers and the intracellular OCN product accumulation under the stimulation of BCP and β-TCP ceramics. Further evidence was found from the formation of mineralized nodules in BCM and TCM. In addition, this study showed a concise relationship between Ca-P ceramic induced inflammation and its osteoinductivity that the increased cytokines and growth factors from macrophages could promote MSCs osteogenic differentiation.
Collapse
Affiliation(s)
- Jing Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xuening Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Bo Guo
- Department of Ophthalmology, West China Hospital of Sichuan University, Chengdu, China
| | - Danyang Li
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| |
Collapse
|
58
|
Gardin C, Bosco G, Ferroni L, Quartesan S, Rizzato A, Tatullo M, Zavan B. Hyperbaric Oxygen Therapy Improves the Osteogenic and Vasculogenic Properties of Mesenchymal Stem Cells in the Presence of Inflammation In Vitro. Int J Mol Sci 2020; 21:ijms21041452. [PMID: 32093391 PMCID: PMC7073059 DOI: 10.3390/ijms21041452] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 02/08/2023] Open
Abstract
Hyperbaric oxygen (HBO) therapy has been reported to be beneficial for treating many conditions of inflammation-associated bone loss. The aim of this work was to in vitro investigate the effect of HBO in the course of osteogenesis of human Mesenchymal Stem Cells (MSCs) grown in a simulated pro-inflammatory environment. Cells were cultured with osteogenic differentiation factors in the presence or not of the pro-inflammatory cytokine Tumor Necrosis Factor-α (TNF-α), and simultaneously exposed daily for 60 min, and up to 21 days, at 2,4 atmosphere absolute (ATA) and 100% O2. To elucidate osteogenic differentiation-dependent effects, cells were additionally pre-committed prior to treatments. Cell metabolic activity was evaluated by means of the MTT assay and DNA content quantification, whereas osteogenic and vasculogenic differentiation was assessed by quantification of extracellular calcium deposition and gene expression analysis. Metabolic activity and osteogenic properties of cells did not differ between HBO, high pressure (HB) alone, or high oxygen (HO) alone and control if cells were pre-differentiated to the osteogenic lineage. In contrast, when treatments started contextually to the osteogenic differentiation of the cells, a significant reduction in cell metabolic activity first, and in mineral deposition at later time points, were observed in the HBO-treated group. Interestingly, TNF-α supplementation determined a significant improvement in the osteogenic capacity of cells subjected to HBO, which was not observed in TNF-α-treated cells exposed to HB or HO alone. This study suggests that exposure of osteogenic-differentiating MSCs to HBO under in vitro simulated inflammatory conditions enhances differentiation towards the osteogenic phenotype, providing evidence of the potential application of HBO in all those processes requiring bone regeneration.
Collapse
Affiliation(s)
- Chiara Gardin
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Gerardo Bosco
- Department of Biomedical Sciences, University of Padova, 35128 Padova, Italy; (G.B.); (S.Q.); (A.R.)
| | - Letizia Ferroni
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Silvia Quartesan
- Department of Biomedical Sciences, University of Padova, 35128 Padova, Italy; (G.B.); (S.Q.); (A.R.)
| | - Alex Rizzato
- Department of Biomedical Sciences, University of Padova, 35128 Padova, Italy; (G.B.); (S.Q.); (A.R.)
| | - Marco Tatullo
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70121 Bari, Italy
- Correspondence: (B.Z.); (M.T.); Tel.: +39-0532-455-502 (B.Z.)
| | - Barbara Zavan
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: (B.Z.); (M.T.); Tel.: +39-0532-455-502 (B.Z.)
| |
Collapse
|
59
|
Lipopolysaccharide (LPS) inhibits ectopic bone formation induced by bone morphogenetic protein-2 and TGF-β1 through IL-1β production. J Oral Biosci 2020; 62:44-51. [PMID: 31987892 DOI: 10.1016/j.job.2020.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 01/06/2023]
Abstract
OBJECTIVES In order to gain new insight into bacterial infection during bone-regenerative treatment using bone morphogenetic proteins (BMPs), we examined the effects of lipopolysaccharide (LPS) on ectopic bone formation induced by BMP-2 and transforming growth factor (TGF)-β1 in mice. METHODS We implanted collagen sponges containing BMP-2, TGF-β1, and various amounts of LPS into mouse muscle tissues. Lump-like masses in which ectopic bones developed in mice were processed for microcomputed tomography, DNA microarray, reverse-transcription PCR, and histological analyses. RESULTS LPS treatment caused a dose-dependent reduction in the volume of ectopic bone. The total volume of ectopic bone induced by BMP-2 + TGF-β1 treatment was reduced by more than 75% in the presence of LPS. Histological analysis of the ectopic bone tissues revealed a significant reduction in total bone volume and bone volume/total volume in response to LPS. LPS treatment significantly increased the osteoblast number and osteoid volume, while the osteoclast number did not change. Since LPS induced production of TNF-α and IL-1β in lump-like masses, we implanted collagen sponges containing BMP-2 and TGF-β1 with or without LPS into TNF-α- or IL-1α/β-deficient mice. LPS treatment reduced the volume of ectopic bones in TNF-α-deficient mice but not in IL-1α/β-deficient mice. Furthermore, collagen sponges containing IL-1β reduced ectopic bone formation by BMP-2 and TGF-β1 in wild-type mice to the same extent as LPS treatment did. CONCLUSIONS LPS suppresses the ectopic bone formation induced by BMP-2 and TGF-β1 through IL-1β production.
Collapse
|
60
|
Qasim M, Chae DS, Lee NY. Bioengineering strategies for bone and cartilage tissue regeneration using growth factors and stem cells. J Biomed Mater Res A 2019; 108:394-411. [PMID: 31618509 DOI: 10.1002/jbm.a.36817] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/03/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022]
Abstract
Bone and cartilage tissue engineering is an integrative approach that is inspired by the phenomena associated with wound healing. In this respect, growth factors have emerged as important moieties for the control and regulation of this process. Growth factors act as mediators and control the important physiological functions of bone regeneration. Herein, we discuss the importance of growth factors in bone and cartilage tissue engineering, their loading and delivery strategies, release kinetics, and their integration with biomaterials and stem cells to heal bone fractures. We also highlighted the role of growth factors in the determination of the bone tissue microenvironment based on the reciprocal signaling with cells and biomaterial scaffolds on which future bone and cartilage tissue engineering technologies and medical devices will be based upon.
Collapse
Affiliation(s)
- Muhammad Qasim
- Department of BioNano Technology, Gachon University, Seongnam-si, Republic of Korea
| | - Dong Sik Chae
- Department of Orthopedic Surgery, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon, Republic of Korea
| | - Nae Yoon Lee
- Department of BioNano Technology, Gachon University, Seongnam-si, Republic of Korea
| |
Collapse
|
61
|
Borem R, Madeline A, Bowman M, Gill S, Tokish J, Mercuri J. Differential Effector Response of Amnion- and Adipose-Derived Mesenchymal Stem Cells to Inflammation; Implications for Intradiscal Therapy. J Orthop Res 2019; 37:2445-2456. [PMID: 31287173 DOI: 10.1002/jor.24412] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/25/2019] [Indexed: 02/04/2023]
Abstract
Intervertebral disc degeneration (IVDD) is a progressive condition marked by tissue destruction and inflammation. The therapeutic effector functions of mesenchymal stem cells (MSCs) makes them an attractive therapy for patients with IVDD. While several sources of MSCs exist, the optimal choice for use in the inflamed IVD remains a significant question. Adipose (AD)- and amnion (AM)-derived MSCs have several advantages compared with other sources, however, no study has directly compared the impact of IVDD inflammation on their effector functions. Human MSCs were cultured in media with or without supplementation of interleukin-1β (IL-1β) and tumor necrosis factor-α at concentrations reportedly produced by IVDD cells. MSC proliferation and production of pro- and anti-inflammatory cytokines were quantified following 24 and 48 h of culture. Additionally, the osteogenic and chondrogenic potential of AD- and AM-MSCs was characterized via histology and biochemical analysis following 28 days of culture. In inflammatory culture, AM-MSCs produced significantly more anti-inflammatory IL-10 (14.47 ± 2.39 pg/ml; p = 0.004) and larger chondrogenic pellets (5.67 ± 0.26 mm2 ; p = 0.04) with greater percent area staining positively for glycosaminoglycan (82.03 ± 3.26%; p < 0.001) compared with AD-MSCs (0.00 ± 0.00 pg/ml; 2.76 ± 0.18 mm2 ; 34.75 ± 2.49%; respectively). Conversely, AD-MSCs proliferated more resulting in higher cell numbers (221,000 ± 8,021 cells; p = 0.048) and produced higher concentrations of pro-inflammatory cytokines prostaglandin E2 (1,118.30 ± 115.56 pg/ml; p = 0.030) and IL-1β (185.40 ± 7.63 pg/ml; p = 0.010) compared with AM-MSCs (109,667 ± 5,696 cells; 1,291.40 ± 78.47 pg/ml; 144.10 ± 4.57 pg/ml; respectively). AD-MSCs produced more mineralized extracellular matrix (3.34 ± 0.05 relative absorbance units [RAU]; p < 0.001) compared with AM-MSCs (1.08 ± 0.06 RAU). Under identical inflammatory conditions, a different effector response was observed with AM-MSCs producing more anti-inflammatories and demonstrating enhanced chondrogenesis compared with AD-MSCs, which produced more pro-inflammatory cytokines and demonstrated enhanced osteogenesis. These findings may begin to help inform researchers which MSC source may be optimal for IVD regeneration. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2445-2456, 2019.
Collapse
Affiliation(s)
- Ryan Borem
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| | - Allison Madeline
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| | - Mackenzie Bowman
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| | - Sanjitpal Gill
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634.,Department of Orthopaedic Surgery, Medical Group of the Carolinas-Pelham, Spartanburg Regional Healthcare System, Greer, South Carolina, 29651
| | - John Tokish
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634.,Department of Orthopaedic Surgery, Mayo Clinic, Phoenix, Arizona, 85054
| | - Jeremy Mercuri
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| |
Collapse
|
62
|
Chen TH, Weber FE, Malina-Altzinger J, Ghayor C. Epigenetic drugs as new therapy for tumor necrosis factor-α-compromised bone healing. Bone 2019; 127:49-58. [PMID: 31152802 DOI: 10.1016/j.bone.2019.05.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/20/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023]
Abstract
Impaired bone regeneration by excess inflammation leads to failure of bone healing. Current therapies display limited benefits making new treatments imperative. Our recent discoveries of the anti-inflammatory characteristics of bromodomain and extra terminal domain (BET) inhibitors, N-methylpyrrolidone (NMP) and N,N-Dimethylacetamide (DMA), implicate possible therapeutic use of epigenetic drugs in inflammation-impaired bone healing. Here, we investigated the effects of NMP and DMA on osteogenesis in vitro and ex vivo under the influence of TNFα, a key cytokine responsible for impaired fracture healing. NMP and DMA pre-treatment recovered TNFα-inhibited expression of essential osteoblastic genes, Alp, Runx2, and Osterix as well as mineralization in multipotent stem cells, but not in pre-osteoblasts and calvarial osteoblasts. The mechanism of action involves the recovery of TNFα-suppressed BMP-induced Smad signaling and the reduction of TNFα-triggered ERK pathway. In addition, ERK inhibitor treatment diminished the effect of TNFα on osteogenesis, which reinforces the role of ERK pathway in the adverse effect of TNFα. Furthermore, endochondral ossification was analyzed in an ex vivo bone culture model. TNFα largely abrogated BMP-promoted growth of mineralized bone while pre-treatment of NMP and DMA prevented the deleterious effect of TNFα. Taken together, these data shed light on developing low- affinity epigenetic drugs as new therapies for inflammation-compromised bone healing.
Collapse
Affiliation(s)
- Tse-Hsiang Chen
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Plattenstrasse11, 8032 Zürich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - Franz E Weber
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Plattenstrasse11, 8032 Zürich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland; CABMM, Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zürich, Switzerland
| | - Johann Malina-Altzinger
- Limmat Cleft and Craniofacial Centre, Zürich MKG, Hardturmstrasse 133, 8005 Zürich, Switzerland
| | - Chafik Ghayor
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Plattenstrasse11, 8032 Zürich, Switzerland.
| |
Collapse
|
63
|
Gonzalez-Fernandez T, Sikorski P, Leach JK. Bio-instructive materials for musculoskeletal regeneration. Acta Biomater 2019; 96:20-34. [PMID: 31302298 PMCID: PMC6717669 DOI: 10.1016/j.actbio.2019.07.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 06/26/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023]
Abstract
The prevalence and cost of disorders affecting the musculoskeletal system are predicted to rise significantly in the coming years due to the aging global population and the increase of associated risk factors. Despite being the second largest cause of disability, the clinical options for therapeutic intervention remain limited. The clinical translation of cell-based therapies for the treatment of musculoskeletal disorders faces many challenges including maintenance of cell survival in the harsh in vivo environment and the lack of control over regulating cell phenotype upon implantation. In order to address these challenges, the development of bio-instructive materials to modulate cell behavior has taken center stage as a strategy to increase the therapeutic potential of various cell populations. However, the determination of the necessary cues for a specific application and how these signals should be presented from a biomaterial remains elusive. This review highlights recent biochemical and physical strategies used to engineer bio-instructive materials for the repair of musculoskeletal tissues. There is a particular emphasis on emerging efforts such as the engineering of immunomodulatory and antibacterial materials, as well as the incorporation of these strategies into biofabrication and organ-on-a-chip approaches. STATEMENT OF SIGNIFICANCE: Disorders affecting the musculoskeletal system affect individuals across the lifespan and have a profound effect on mobility and quality of life. While small defects in many tissues can heal successfully, larger defects are often unable to heal or instead heal with inferior quality fibrous tissue and require clinical intervention. Cell-based therapies are a promising option for clinical translation, yet challenges related to maintaining cell survival and instructing cell phenotype upon implantation have limited the success of this approach. Bio-instructive materials provide an exciting opportunity to modulate cell behavior and enhance the efficacy of cell-based approaches for musculoskeletal repair. However, the identification of critical instructive cues and how to present these stimuli is a focus of intense investigation. This review highlights recent biochemical and physical strategies used to engineer bio-instructive materials for the repair of musculoskeletal tissues, while also considering exciting progress in the engineering of immunomodulatory and antibacterial materials.
Collapse
Affiliation(s)
| | - Pawel Sikorski
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA; Department of Physics, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA; Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA.
| |
Collapse
|
64
|
Sadowska JM, Wei F, Guo J, Guillem-Marti J, Lin Z, Ginebra MP, Xiao Y. The effect of biomimetic calcium deficient hydroxyapatite and sintered β-tricalcium phosphate on osteoimmune reaction and osteogenesis. Acta Biomater 2019; 96:605-618. [PMID: 31269454 DOI: 10.1016/j.actbio.2019.06.057] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/25/2019] [Accepted: 06/27/2019] [Indexed: 11/15/2022]
Abstract
Biomaterial implantation triggers inflammatory reactions. Understanding the effect of physicochemical features of biomaterials on the release of inflammatory cytokines from immune cells would be of great interest in view of designing bone graft materials to enhance the healing of bone defects. The present work investigated the interactions of two chemically and texturally different calcium phosphate (CaPs) substrates with macrophages, one of the main innate immune cells, and its further impact on osteogenic differentiation of bone forming cells. The behaviour of macrophages seeded on biomimetic calcium deficient hydroxyapatite (CDHA) and sintered β-tricalcium phosphate (β-TCP) was assessed in terms of the release of inflammatory cytokines and osteoclastogenic factors. The osteogenic differentiation of bone progenitor cells (bone marrow stromal cells (BMSCs) and osteoblastic cell line (SaOS-2)) were subsequently studied by incubating with the conditioned medium induced by macrophage-CaPs interaction in order to reveal the effect of immune cell reaction to CaPs on osteogenic differentiation. It was found that the incubation of macrophages with CaPs substrates caused a decrease of pro-inflammatory cytokines, more pronounced for β-TCP compared with CDHA showing significantly decreased IL-6, TNF-a, and iNOS. However, the macrophage-CDHA interaction resulted in a more favourable environment for osteogenic differentiation of osteoblasts with more collagen type I production and osteogenic genes (Runx2, BSP) expression, suggesting that osteogenic differentiation of bone cells is not only determined by the nature of biomaterials, but also significantly influenced by the inflammatory environment generated by the interaction of immune cells and biomaterials. STATEMENT OF SIGNIFICANCE: The field of osteoimmunology highlights the importance of the cross-talk between immune and bone cells for effective bone regeneration. This tight interaction opens the door to new strategies that encompass the development of smart cell-instructive biomaterials which performance covers the events from early inflammation to osteogenesis. The present work links the anti-inflammatory and osteoimmunomodulatory features of synthetic bone grafts to their chemistry and texture, focussing on the cross-talk between macrophages and two major orchestrators of bone healing, namely primary mesenchymal stem cells and osteoblasts. The results emphasize the importance of the microenvironment created through the interaction between the substrate and the immune cells as it can stimulate osteogenic events and subsequently foster bone healing.
Collapse
Affiliation(s)
- Joanna M Sadowska
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain; Barcelona Research Centre in Multiscale Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain.
| | - Fei Wei
- Institute of Health and Biomedical Innovation and the Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, QLD 4059, Australia.
| | - Jia Guo
- Institute of Health and Biomedical Innovation and the Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, QLD 4059, Australia; Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guanghua Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510055, People's Republic of China.
| | - Jordi Guillem-Marti
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain; Barcelona Research Centre in Multiscale Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain.
| | - Zhengmei Lin
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University and Guanghua Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510055, People's Republic of China.
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain; Barcelona Research Centre in Multiscale Science and Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany 16, 08019 Barcelona, Spain; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain.
| | - Yin Xiao
- Institute of Health and Biomedical Innovation and the Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, QLD 4059, Australia.
| |
Collapse
|
65
|
Li L, Tang P, Zhou Z, Wang Q, Xu T, Zhao S, Huang Y, Kong F, Liu W, Cheng L, Zhou Z, Zhao X, Gu C, Luo Y, Tao G, Qian D, Chen J, Fan J, Yin G. GIT1 regulates angiogenic factor secretion in bone marrow mesenchymal stem cells via NF-κB/Notch signalling to promote angiogenesis. Cell Prolif 2019; 52:e12689. [PMID: 31502302 PMCID: PMC6869488 DOI: 10.1111/cpr.12689] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/23/2019] [Accepted: 08/11/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives Osteogenesis is coupled with angiogenesis during bone remodelling. G‐protein‐coupled receptor (GPCR) kinase 2‐interacting protein‐1 (GIT1) is an important protein that participates in fracture healing by regulating angiogenesis. This study investigated whether GIT1 could affect bone mesenchymal stem cells (BMSCs) to secrete angiogenic factors to enhance fracture healing by promoting angiogenesis and its possible mechanism. Materials and methods The angiogenesis of mice post‐fracture was detected by micro‐CT and immunofluorescence. Subsequently, vascular endothelial growth factor (VEGF) level in mouse and human BMSCs (hBMSCs) under TNF‐α stimulation was detected. The hBMSCs were transfected with GIT1 shRNAs to further explore the relationship between GIT1 and VEGF and angiogenesis in vitro. Furthermore, based on previous research on GIT1, possible signal pathways were investigated. Results GIT1 knockout mice exhibited impaired angiogenesis and delayed fracture healing. And GIT1 deficiency remarkably reduced the expression of VEGF mRNA in BMSCs, which affected the proliferation and migration of human umbilical vein endothelial cells. GIT1 knockdown inhibited the activation of Notch and NF‐κB signals by decreasing nuclear transportation of NICD and P65/P50, respectively. Overexpression of the canonical NF‐κB subunits P65 and P50 markedly increased NICD‐dependent activation of recombination signal‐binding protein‐jκ reporter. Finally, GIT1 enhanced the affinity of NF‐κB essential modulator (NEMO) for K63‐linked ubiquitin chains via interaction with NEMO coiled‐coil 2 domains. Conclusion These data revealed a positive role for GIT1 by modulating the Notch/NF‐κB signals which promoting paracrine of BMSCs to enhance angiogenesis and fracture healing.
Collapse
Affiliation(s)
- Linwei Li
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pengyu Tang
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Zhou
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Wang
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Xu
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shujie Zhao
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifan Huang
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fanqi Kong
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Liu
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Cheng
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhimin Zhou
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan Zhao
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Changjiang Gu
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongjun Luo
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gaojian Tao
- Department of Pain, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Dingfei Qian
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Chen
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Fan
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guoyong Yin
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
66
|
Interleukin-35 stimulates tumor necrosis factor-α activated osteoblasts differentiation through Wnt/β-catenin signaling pathway in rheumatoid arthritis. Int Immunopharmacol 2019; 75:105810. [PMID: 31404890 DOI: 10.1016/j.intimp.2019.105810] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/31/2019] [Accepted: 08/05/2019] [Indexed: 02/08/2023]
Abstract
Interleukin (IL)-35 plays an important role in the pathogenesis of rheumatoid arthritis (RA), which is characterized by tumor necrosis factor (TNF)-α activated bone loss beginning early and persisting over time. The aim of this study was to explore the effects and signaling pathway of IL-35 on osteoblasts differentiation in MC3T3E1 cells and TNF-α activated MC3T3E1 cells. A microenvironment was established with low concentration and short-term treatment of TNF-α to mimic inflammatory activated osteoblasts of RA in vitro. The role of IL-35 on osteoblasts proliferation and apoptosis were assessed using cell counting kit (CCK)-8 assay and flow cytometry, respectively. Alkaline phosphatase (ALP) activity was measured by p-nitrophenyl phosphate assay. Extracellular matrix mineralization was measured by Alizarin red S staining. Osteoprotegerin (OPG) and receptor activator of nuclear factor-κB ligand (RANKL) in response to IL-35 were investigated using real-time polymerase chain reaction and western blot analysis. Wnt/β-catenin signaling pathway in osteoblasts was investigated. In basal and TNF-α activated osteoblasts, IL-35 promoted proliferation and inhibited apoptosis. Basal and TNF-α activated ALP activity and mineralization in vitro was increased stimulated by IL-35. Furthermore, IL-35 increased the basal and TNF-α activated OPG expression and decreased basal and TNF-α activated RANKL expression. Blocking Wnt/β-catenin signaling pathway with Dickkopf (Dkk)-1 inhibited the osteogenic effects of IL-35. IL-35 stimulates basal and TNF-α activated osteoblasts differentiation through the Wnt/β-catenin signaling pathway, thus highlighting the IL-35 for pharmaceutical and medicinal applications for treating RA bone loss.
Collapse
|
67
|
Wan W, Cheng B, Zhang C, Ma Y, Li A, Xu F, Lin M. Synergistic Effect of Matrix Stiffness and Inflammatory Factors on Osteogenic Differentiation of MSC. Biophys J 2019; 117:129-142. [PMID: 31178039 PMCID: PMC6626830 DOI: 10.1016/j.bpj.2019.05.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/18/2019] [Accepted: 05/09/2019] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) in vivo reside in a complex microenvironment. Changes of both biochemical and biophysical cues in the microenvironment caused by inflammation affect the differentiation behaviors of MSCs. Most studies, however, only focus on either biochemical or biophysical cues, although the synergistic effect of matrix stiffness and inflammatory factors on osteogenic differentiation of MSCs has not been explored yet. Here, we showed that there was a matrix stiffness-dependent modulation in the osteogenic differentiation of human MSCs (hMSCs) with higher matrix stiffness favoring osteogenesis bias. However, when interleukin-1 β (IL-1β) was added, the osteogenic differentiation of hMSCs was suppressed, which was independent of increasing matrix stiffness. Both experimental observations and mathematical modeling confirmed that matrix stiffness and IL-1β could activate the ERK1/2 signaling and contribute to osteogenic differentiation. The p38 signaling activated by IL-1β has a strong role in inhibiting osteoblastic differentiation, thus diminishing the vital effect of ERK1/2 signaling. In addition, sensitivity analysis of the model parameters revealed that activation/deactivation dynamics of sensitive factors (e.g., FAK, ERK, and p38) also played a key role in the synergistic effect of matrix stiffness and IL-1β on the osteogenic differentiation of hMSCs. The outcomes of this study provide new insights into the synergistic effect of biochemical and biophysical microenvironments on regulating MSC differentiation.
Collapse
Affiliation(s)
- Wanting Wan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology; Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Cheng Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yufei Ma
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an, P.R. China.
| |
Collapse
|
68
|
Xiao L, Zhou Y, Friis T, Beagley K, Xiao Y. S1P-S1PR1 Signaling: the "Sphinx" in Osteoimmunology. Front Immunol 2019; 10:1409. [PMID: 31293578 PMCID: PMC6603153 DOI: 10.3389/fimmu.2019.01409] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/04/2019] [Indexed: 12/24/2022] Open
Abstract
The fundamental interaction between the immune and skeletal systems, termed as osteoimmunology, has been demonstrated to play indispensable roles in the maintenance of balance between bone resorption and formation. The pleiotropic sphingolipid metabolite, sphingosine 1-phosphate (S1P), together with its cognate receptor, sphingosine-1-phosphate receptor-1 (S1PR1), are known as key players in osteoimmunology due to the regulation on both immune system and bone remodeling. The role of S1P-S1PR1 signaling in bone remodeling can be directly targeting both osteoclastogenesis and osteogenesis. Meanwhile, inflammatory cell function and polarization in both adaptive immune (T cell subsets) and innate immune cells (macrophages) are also regulated by this signaling axis, suggesting that S1P-S1PR1 signaling could aslo indirectly regulate bone remodeling via modulating the immune system. Therefore, it could be likely that S1P-S1PR1 signaling might take part in the maintenance of continuous bone turnover under physiological conditions, while lead to the pathogenesis of bone deformities during inflammation. In this review, we summarized the immunological regulation of S1P-S1PR1 signal axis during bone remodeling with an emphasis on how osteo-immune regulators are affected by inflammation, an issue with relevance to chronical bone disorders such as rheumatoid arthritis, spondyloarthritis and periodontitis.
Collapse
Affiliation(s)
- Lan Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, Australia
| | - Yinghong Zhou
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, Australia.,Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Thor Friis
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kenneth Beagley
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, Australia
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,The Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, Australia.,Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
69
|
Li X, Li Z, Wang J, Li Z, Cui H, Dai G, Chen S, Zhang M, Zheng Z, Zhan Z, Liu H. Wnt4 signaling mediates protective effects of melatonin on new bone formation in an inflammatory environment. FASEB J 2019; 33:10126-10139. [PMID: 31216173 DOI: 10.1096/fj.201900093rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Growing evidence shows that the inhibitory effect of inflammatory cytokines on new bone formation by osteogenic precursor cells is a critical cause of net bone-density reduction. Melatonin has been proven to be a potential therapeutic candidate for osteoporosis. However, whether it is capable of antagonizing the suppressing effect of inflammatory cytokines on osteogenic precursor cells is so far elusive. In this study, using the cell culture system of human bone marrow stromal cells and MC3T3-E1 preosteoblasts, we recorded the following vital observations that provided insights of melatonin-induced bone formation: 1) melatonin induced bone formation in both normal and inflammatory conditions; 2) Wnt4 was essential for melatonin-induced bone formation in inflammatory stimulation; 3) melatonin- and Wnt4-induced bone formation occurred via activation of β-catenin and p38-JNK MAPK pathways by interaction with a distinct frizzled LDL receptor-related protein complex; 4) melatonin suppressed the inhibitory effect of NF-κB on osteogenesis in a Wnt4-dependent manner; and 5) melatonin induced Wnt4 expression through the ERK1/2-Pax2-Egr1 pathway. In summary, we showed a novel mechanism of melatonin-induced bone formation in an inflammatory environment. Melatonin-induced Wnt4 expression is essential for its osteoinductive effect and the inhibitory effect of NF-κB on bone formation. Our novel findings may provide useful information for its potential translational application.-Li, X., Li, Z., Wang, J., Li, Z., Cui, H., Dai, G., Chen, S., Zhang, M., Zheng, Z., Zhan, Z., Liu, H. Wnt4 signaling mediates protective effects of melatonin on new bone formation in an inflammatory environment.
Collapse
Affiliation(s)
- Xiang Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zihao Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianru Wang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Zemin Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Haowen Cui
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guo Dai
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Siwen Chen
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mingliang Zhang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaomin Zheng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Zhongping Zhan
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Liu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| |
Collapse
|
70
|
Zhang T, Yao Y. Effects of inflammatory cytokines on bone/cartilage repair. J Cell Biochem 2019; 120:6841-6850. [PMID: 30335899 DOI: 10.1002/jcb.27953] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/02/2018] [Indexed: 01/24/2023]
Abstract
Many inflammatory factors can affect cell behaviors and work as a form of inter-regulatory networks through the inflammatory pathway. Inflammatory cytokines are critical for triggering bone regeneration after fracture or bone injury. Also, inflammatory cytokines play an important role in cartilage repair. The synergistic or antagonistic effects of both proinflammatory and anti-inflammatory cytokines have a great influence on fracture healing. This review discusses key inflammatory cytokines and signaling pathways involved in bone or cartilage repair.
Collapse
Affiliation(s)
- Tingshuai Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology And Implant Materials, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongchang Yao
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology And Implant Materials, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
71
|
Toosi S, Naderi-Meshkin H, Kalalinia F, HosseinKhani H, Heirani-Tabasi A, Havakhah S, Nekooei S, Jafarian AH, Rezaie F, Peivandi MT, Mesgarani H, Behravan J. Bone defect healing is induced by collagen sponge/polyglycolic acid. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2019; 30:33. [PMID: 30840143 DOI: 10.1007/s10856-019-6235-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 02/11/2019] [Indexed: 06/09/2023]
Abstract
We have evaluated the capability of a collagen/poly glycolic acid (PGA) scaffold in regeneration of a calvarial bone defects in rabbits. 4 bone critical size defects (CSD) were created in the calvarial bone of each rabbit. The following 4 treatment modalities were tested (1) a collagen/PGA scaffold (0.52% w/w); (2) the collagen/PGA scaffold (0.52% w/w) seeded with adipose-derived mesenchymal stem cells (AD-MSCs, 1 × 106 cells per each defect); (3) AD-MSCs (1 × 106 cells) no scaffold material, and (4) blank control. The rabbits were then divided into 3 random groups (of 5) and the treatment outcomes were evaluated at 4, 8 and 12 weeks. New bone formation was histologically assessed. Experimental groups were analyzed by CT scan and real-time PCR. Histological analysis of bone defects treated with collagen/PGA alone exhibited significant fibrous connective tissue formation at the 12 weeks of treatments (P ≤ 0.05). There was no significant difference between collagen/PGA alone and collagen/PGA + AD-MSCs groups. The results were confirmed by CT scan data showing healing percentages of 34.20% for the collage/PGA group alone as compared to the control group and no difference with collagen/PGA containing AD-MSCs (1 × 106 cells). RT-PCR analysis also indicated no significant differences between collagen/PGA and collagen/PGA + AD-MSC groups, although both scaffold containing groups significantly express ALP and SIO rather than groups without scaffolds. Although there was no significant difference between the scaffolds containing cells with non-cellular scaffolds, our results indicated that the Collagen/PGA scaffold itself had a significant effect on wound healing as compared to the control group. Therefore, the collagen/PGA scaffold seems to be a promising candidate for research in bone regeneration.
Collapse
Affiliation(s)
- Shirin Toosi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- NanoSBY Knowledge Based Corporation, Mashhad, Iran
| | - Hojjat Naderi-Meshkin
- NanoSBY Knowledge Based Corporation, Mashhad, Iran
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Fatemeh Kalalinia
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein HosseinKhani
- Innovation Center for Advanced Technology, Matrix, Inc., New York, NY, 10029, USA
| | - Asieh Heirani-Tabasi
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Shahrzad Havakhah
- Physiology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | - Fahimeh Rezaie
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Mohammad Taghi Peivandi
- Department of Orthopedic Surgery, Orthopedic and Trauma Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hooman Mesgarani
- Department of Veterinary Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Javad Behravan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- NanoSBY Knowledge Based Corporation, Mashhad, Iran.
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.
- Center for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
72
|
Xiao L, Xiao Y. The Autophagy in Osteoimmonology: Self-Eating, Maintenance, and Beyond. Front Endocrinol (Lausanne) 2019; 10:490. [PMID: 31428045 PMCID: PMC6689986 DOI: 10.3389/fendo.2019.00490] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 07/08/2019] [Indexed: 12/12/2022] Open
Abstract
It has been long realized that the immune and skeletal systems are closely linked. This crosstalk, also known as osteoimmunology, is a primary process required for bone health. For example, the immune system acts as a key regulator in osteoclasts-osteoblasts coupling to maintain the balanced bone remodeling. Osteoimmunology is achieved through many cellular and molecular processes, among which autophagy has recently been found to play an indispensable role. Autophagy is a highly conserved process in eukaryotic cells, by which the cytoplasm components such as dysfunctional organelles are degraded through lysosomes and then returned to the cytosol for reuse. Autophagy is present in all cells at basal levels to maintain homeostasis and to promote cell survival in response to cellular stress conditions such as nutrition deprivation and hypoxia. Autophagy is a required process in immune cell activation/polarization and osteoclast differentiation, which protecting cells from oxidative stress. The essential of autophagy in osteogenesis is its involvement in osteoblast differentiation and mineralization, especially the role of autophagosome in extracellular calcium transportation. The modulatory feature of autophagy in both immune and skeleton systems suggests its crucial roles in osteoimmunology. Furthermore, autophagy also participates in the maintenance of bone marrow hematopoietic stem cell niche. The focus of this review is to highlight the role of autophagy in the immune-skeleton interactions and the effects on bone physiology, as well as the future application in translational research.
Collapse
Affiliation(s)
- Lan Xiao
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, QLD, Australia
| | - Yin Xiao
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, QLD, Australia
- *Correspondence: Yin Xiao
| |
Collapse
|
73
|
Mödinger Y, Rapp AE, Vikman A, Ren Z, Fischer V, Bergdolt S, Haffner-Luntzer M, Song WC, Lambris JD, Huber-Lang M, Neidlinger-Wilke C, Brenner RE, Ignatius A. Reduced Terminal Complement Complex Formation in Mice Manifests in Low Bone Mass and Impaired Fracture Healing. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:147-161. [PMID: 30339839 DOI: 10.1016/j.ajpath.2018.09.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/01/2018] [Accepted: 09/17/2018] [Indexed: 12/20/2022]
Abstract
The terminal complement complex (TCC) is formed on activation of the complement system, a crucial arm of innate immunity. TCC formation on cell membranes results in a transmembrane pore leading to cell lysis. In addition, sublytic TCC concentrations can modulate various cellular functions. TCC-induced effects may play a role in the pathomechanisms of inflammatory disorders of the bone, including rheumatoid arthritis and osteoarthritis. In this study, we investigated the effect of the TCC on bone turnover and repair. Mice deficient for complement component 6 (C6), an essential component for TCC assembly, and mice with a knockout of CD59, which is a negative regulator of TCC formation, were used in this study. The bone phenotype was analyzed in vivo, and bone cell behavior was analyzed ex vivo. In addition, the mice were subjected to a femur osteotomy. Under homeostatic conditions, C6-deficient mice displayed a reduced bone mass, mainly because of increased osteoclast activity. After femur fracture, the inflammatory response was altered and bone formation was disturbed, which negatively affected the healing outcome. By contrast, CD59-knockout mice only displayed minor skeletal alterations and uneventful bone healing, although the early inflammatory reaction to femur fracture was marginally enhanced. These results demonstrate that TCC-mediated effects regulate bone turnover and promote an adequate response to fracture, contributing to an uneventful healing outcome.
Collapse
Affiliation(s)
- Yvonne Mödinger
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anna E Rapp
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anna Vikman
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Zhaozhou Ren
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Stephanie Bergdolt
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Wen-Chao Song
- Department of Pharmacology and Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University Medical Center, Ulm, Germany
| | | | - Rolf E Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Trauma Research Center Ulm, Ulm University Medical Center, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
74
|
Sayardoust S, Omar O, Norderyd O, Thomsen P. Implant-associated gene expression in the jaw bone of smokers and nonsmokers: A human study using quantitative qPCR. Clin Oral Implants Res 2018; 29:937-953. [DOI: 10.1111/clr.13351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 07/04/2018] [Accepted: 07/10/2018] [Indexed: 01/11/2023]
Affiliation(s)
- Shariel Sayardoust
- Department of Biomaterials; Institute of Clinical Sciences; Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
- Department of Periodontology; Institute for Postgraduate Dental Education; Jönköping Sweden
| | - Omar Omar
- Department of Biomaterials; Institute of Clinical Sciences; Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
| | - Ola Norderyd
- Department of Periodontology; Institute for Postgraduate Dental Education; Jönköping Sweden
| | - Peter Thomsen
- Department of Biomaterials; Institute of Clinical Sciences; Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
| |
Collapse
|
75
|
Effect of nano-structural properties of biomimetic hydroxyapatite on osteoimmunomodulation. Biomaterials 2018; 181:318-332. [PMID: 30098568 DOI: 10.1016/j.biomaterials.2018.07.058] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/22/2018] [Accepted: 07/28/2018] [Indexed: 12/27/2022]
Abstract
Immune cells are sensitive to the microstructural and textural properties of materials. Tuning the structural features of synthetic bone grafts could be a valuable strategy to regulate the specific response of the immune system, which in turn modulates the activity of bone cells. The aim of this study was to analyse the effect of the structural characteristics of biomimetic calcium deficient hydroxyapatite (CDHA) on the innate immune response of macrophages and the subsequent impact on osteogenesis and osteoclastogenesis. Murine RAW 264.7 cells were cultured, under standard and inflammatory conditions, on chemically identical CDHA substrates that varied in microstructure and porosity. The impact on osteogenesis was evaluated by incubating osteoblastic cells (SaOS-2) with RAW-CDHA conditioned extracts. The results showed that macrophages were sensitive to different textural and structural properties of CDHA. Under standard conditions, the impact of inflammatory cytokine production by RAW cells cultured on CDHA played a significant role in the degradation of substrates, suggesting the impact of resorptive behaviour of RAW cells on biomimetic surfaces. Osteoblast differentiation was stimulated by the conditioned media collected from RAW cells cultured on needle-like nanostructured CDHA. The results demonstrated that needle-like nanostructured CDHA was able to generate a favourable osteoimmune environment to regulate osteoblast differentiation and osteogenesis. Under inflammatory conditions, the incubation of RAW cells with less porous CDHA resulted in a decreased gene expression and release of pro-inflammatory cytokines.
Collapse
|
76
|
Bottini M, Mebarek S, Anderson KL, Strzelecka-Kiliszek A, Bozycki L, Simão AMS, Bolean M, Ciancaglini P, Pikula JB, Pikula S, Magne D, Volkmann N, Hanein D, Millán JL, Buchet R. Matrix vesicles from chondrocytes and osteoblasts: Their biogenesis, properties, functions and biomimetic models. Biochim Biophys Acta Gen Subj 2018; 1862:532-546. [PMID: 29108957 PMCID: PMC5801150 DOI: 10.1016/j.bbagen.2017.11.005] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 10/28/2017] [Accepted: 11/01/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Matrix vesicles (MVs) are released from hypertrophic chondrocytes and from mature osteoblasts, the cells responsible for endochondral and membranous ossification. Under pathological conditions, they can also be released from cells of non-skeletal tissues such as vascular smooth muscle cells. MVs are extracellular vesicles of approximately 100-300nm diameter harboring the biochemical machinery needed to induce mineralization. SCOPE OF THE REVIEW The review comprehensively delineates our current knowledge of MV biology and highlights open questions aiming to stimulate further research. The review is constructed as a series of questions addressing issues of MVs ranging from their biogenesis and functions, to biomimetic models. It critically evaluates experimental data including their isolation and characterization methods, like lipidomics, proteomics, transmission electron microscopy, atomic force microscopy and proteoliposome models mimicking MVs. MAJOR CONCLUSIONS MVs have a relatively well-defined function as initiators of mineralization. They bind to collagen and their composition reflects the composition of lipid rafts. We call attention to the as yet unclear mechanisms leading to the biogenesis of MVs, and how minerals form and when they are formed. We discuss the prospects of employing upcoming experimental models to deepen our understanding of MV-mediated mineralization and mineralization disorders such as the use of reconstituted lipid vesicles, proteoliposomes and, native sample preparations and high-resolution technologies. GENERAL SIGNIFICANCE MVs have been extensively investigated owing to their roles in skeletal and ectopic mineralization. MVs serve as a model system for lipid raft structures, and for the mechanisms of genesis and release of extracellular vesicles.
Collapse
Affiliation(s)
- Massimo Bottini
- University of Rome Tor Vergata, Department of Experimental Medicine and Surgery, 00133 Roma, Italy; Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Saida Mebarek
- Universite Lyon 1, UFR Chimie Biochimie, 69 622 Villeurbanne Cedex, France; ICBMS UMR 5246 CNRS, 69 622 Villeurbanne Cedex, France; INSA, Lyon, 69 622 Villeurbanne Cedex, France; CPE, Lyon, 69 622 Villeurbanne Cedex, France; Universite de Lyon, 69 622 Villeurbanne Cedex, France
| | - Karen L Anderson
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Agnieszka Strzelecka-Kiliszek
- Nencki Institute of Experimental Biology, Department of Biochemistry, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Lukasz Bozycki
- Nencki Institute of Experimental Biology, Department of Biochemistry, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Ana Maria Sper Simão
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, USP, Departamento de Química, 14040-901 Ribeirão Preto, SP, Brazil
| | - Maytê Bolean
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, USP, Departamento de Química, 14040-901 Ribeirão Preto, SP, Brazil
| | - Pietro Ciancaglini
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, USP, Departamento de Química, 14040-901 Ribeirão Preto, SP, Brazil
| | - Joanna Bandorowicz Pikula
- Nencki Institute of Experimental Biology, Department of Biochemistry, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Slawomir Pikula
- Nencki Institute of Experimental Biology, Department of Biochemistry, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - David Magne
- Universite Lyon 1, UFR Chimie Biochimie, 69 622 Villeurbanne Cedex, France; ICBMS UMR 5246 CNRS, 69 622 Villeurbanne Cedex, France; INSA, Lyon, 69 622 Villeurbanne Cedex, France; CPE, Lyon, 69 622 Villeurbanne Cedex, France; Universite de Lyon, 69 622 Villeurbanne Cedex, France
| | - Niels Volkmann
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Dorit Hanein
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - José Luis Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Rene Buchet
- Universite Lyon 1, UFR Chimie Biochimie, 69 622 Villeurbanne Cedex, France; ICBMS UMR 5246 CNRS, 69 622 Villeurbanne Cedex, France; INSA, Lyon, 69 622 Villeurbanne Cedex, France; CPE, Lyon, 69 622 Villeurbanne Cedex, France; Universite de Lyon, 69 622 Villeurbanne Cedex, France.
| |
Collapse
|
77
|
Rios Rodriguez V, Llop M, Poddubnyy D. Hematopoietic and mesenchymal stem cells: a promising new therapy for spondyloarthritis? Immunotherapy 2018; 9:899-911. [PMID: 29338611 DOI: 10.2217/imt-2017-0034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In the last years, a considerable progress has been made in the treatment of spondyloarthritides. Nonetheless, there remain a considerable number of patients who are unresponsive to all current therapies. Since the late 1990s, numerous trials have investigated the use of stem cell transplantation as a new approach for the treatment of autoimmune disease, particularly with hematopoietic stem cell transplantation. More recently, the research has focused on mesenchymal stem cell application due to their low immunogenicity and immunomodulatory properties. In this article, we summarize available data on hematopoietic stem cell and mesenchymal stem cell use for the treatment of spondyloarthritides and discuss the data gaps and possible research agenda in this area.
Collapse
Affiliation(s)
- Valeria Rios Rodriguez
- Department of Gastroenterology, Infectiology & Rheumatology, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Llop
- Department of Rheumatology, Hospital Ramón y Cajal, Madrid, Spain
| | - Denis Poddubnyy
- Department of Gastroenterology, Infectiology & Rheumatology, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
78
|
Wei F, Xiao Y. Modulation of the Osteoimmune Environment in the Development of Biomaterials for Osteogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1077:69-86. [DOI: 10.1007/978-981-13-0947-2_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
79
|
Shu Y, Yu Y, Zhang S, Wang J, Xiao Y, Liu C. The immunomodulatory role of sulfated chitosan in BMP-2-mediated bone regeneration. Biomater Sci 2018; 6:2496-2507. [DOI: 10.1039/c8bm00701b] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The immunomodulatory property of biomaterials is vital in determining the in vivo fate of implants and tissue regeneration.
Collapse
Affiliation(s)
- Yang Shu
- Key Laboratory for Ultrafine Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- PR China
- Engineering Research Center for Biomaterials of Ministry of Education
| | - Yuanman Yu
- Key Laboratory for Ultrafine Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- PR China
- Engineering Research Center for Biomaterials of Ministry of Education
| | - Shuang Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- PR China
- Engineering Research Center for Biomaterials of Ministry of Education
| | - Jing Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- PR China
- Engineering Research Center for Biomaterials of Ministry of Education
| | - Yin Xiao
- Australia-China Center for Tissue Engineering and Regenerative Medicine
- Queensland University of Technology
- Brisbane
- Australia
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai 200237
- PR China
- Engineering Research Center for Biomaterials of Ministry of Education
| |
Collapse
|
80
|
Krajewska-Włodarczyk M, Owczarczyk-Saczonek A, Placek W, Osowski A, Engelgardt P, Wojtkiewicz J. Role of Stem Cells in Pathophysiology and Therapy of Spondyloarthropathies-New Therapeutic Possibilities? Int J Mol Sci 2017; 19:ijms19010080. [PMID: 29283375 PMCID: PMC5796030 DOI: 10.3390/ijms19010080] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 12/23/2017] [Accepted: 12/25/2017] [Indexed: 12/14/2022] Open
Abstract
Considerable progress has been made recently in understanding the complex pathogenesis and treatment of spondyloarthropathies (SpA). Currently, along with traditional disease modifying anti-rheumatic drugs (DMARDs), TNF-α, IL-12/23 and IL-17 are available for treatment of such diseases as ankylosing spondylitis (AS) and psoriatic arthritis (PsA). Although they adequately control inflammatory symptoms, they do not affect the abnormal bone formation processes associated with SpA. However, the traditional therapeutic approach does not cover the regenerative treatment of damaged tissues. In this regards, stem cells may offer a promising, safe and effective therapeutic option. The aim of this paper is to present the role of mesenchymal stromal cells (MSC) in pathogenesis of SpA and to highlight the opportunities for using stem cells in regenerative processes and in the treatment of inflammatory changes in articular structures.
Collapse
Affiliation(s)
- Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, Municipal Hospital in Olsztyn, 10-900 Olsztyn, Poland.
- Department of Pathophysiology, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Waldemar Placek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Adam Osowski
- Department of Pathophysiology, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Piotr Engelgardt
- Department of Forensic Medicine, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Laboratory for Regenerative Medicine, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Foundation for Nerve Cell Regeneration, University of Warmia and Mazury in Olsztyn, 10-900 Olsztyn, Poland.
| |
Collapse
|
81
|
Fakhry M, Skafi N, Fayyad-Kazan M, Kobeissy F, Hamade E, Mebarek S, Habib A, Borghol N, Zeidan A, Magne D, Fayyad-Kazan H, Badran B. Characterization and assessment of potential microRNAs involved in phosphate-induced aortic calcification. J Cell Physiol 2017; 233:4056-4067. [PMID: 28776684 DOI: 10.1002/jcp.26121] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/01/2017] [Indexed: 02/01/2023]
Abstract
Medial artery calcification, a hallmark of type 2 diabetes mellitus and chronic kidney disease (CKD), is known as an independent risk factor for cardiovascular mortality and morbidity. Hyperphosphatemia associated with CKD is a strong stimulator of vascular calcification but the molecular mechanisms regulating this process remain not fully understood. We showed that calcification was induced after exposing Sprague-Dawley rat aortic explants to high inorganic phosphate level (Pi , 6 mM) as examined by Alizarin red and Von Kossa staining. This calcification was associated with high Tissue-Nonspecific Alkaline Phosphatase (TNAP) activity, vascular smooth muscle cells de-differentiation, manifested by downregulation of smooth muscle 22 alpha (SM22α) protein expression which was assessed by immunoblot analysis, immunofluorescence, and trans-differentiation into osteo-chondrocyte-like cells revealed by upregulation of Runt related transcription factor 2 (Runx2), TNAP, osteocalcin, and osteopontin mRNA levels which were determined by quantitative real-time PCR. To unravel the possible mechanism(s) involved in this process, microRNA (miR) expression profile, which was assessed using TLDA technique and thereafter confirmed by individual qRT-PCR, revealed differential expression 10 miRs, five at day 3 and 5 at day 6 post Pi treatment versus control untreated aortas. At day 3, miR-200c, -155, 322 were upregulated and miR-708 and 331 were downregulated. After 6 days of treatment, miR-328, -546, -301a were upregulated while miR-409 and miR-542 were downregulated. Our results indicate that high Pi levels trigger aortic calcification and modulation of certain miRs. These observations suggest that mechanisms regulating aortic calcification might involve miRs, which warrant further investigations in future studies.
Collapse
Affiliation(s)
- Maya Fakhry
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon.,Institute of Molecular and Supramolecular Chemistry and Biochemistry (ICBMS), UMR CNRS 5246, University of Lyon 1, Bâtiment Raulin, Villeurbanne Cedex, France
| | - Najwa Skafi
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon.,Institute of Molecular and Supramolecular Chemistry and Biochemistry (ICBMS), UMR CNRS 5246, University of Lyon 1, Bâtiment Raulin, Villeurbanne Cedex, France
| | - Mohammad Fayyad-Kazan
- Institut de Biologie et de Médecine Moléculaires, Université Libre de Bruxelles, Gosselies, Belgium
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Eva Hamade
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| | - Saida Mebarek
- Institute of Molecular and Supramolecular Chemistry and Biochemistry (ICBMS), UMR CNRS 5246, University of Lyon 1, Bâtiment Raulin, Villeurbanne Cedex, France
| | - Aida Habib
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, and the Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, Faculté de Médecine, Site Xavier Bichat, Université Paris Diderot, Paris, France
| | - Nada Borghol
- Institute of Molecular and Supramolecular Chemistry and Biochemistry (ICBMS), UMR CNRS 5246, University of Lyon 1, Bâtiment Raulin, Villeurbanne Cedex, France
| | - Asad Zeidan
- Cardiovascular Physiology Lab, Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,College of Medicine, Qatar University, Doha, Qatar
| | - David Magne
- Institute of Molecular and Supramolecular Chemistry and Biochemistry (ICBMS), UMR CNRS 5246, University of Lyon 1, Bâtiment Raulin, Villeurbanne Cedex, France
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| |
Collapse
|
82
|
IL-1/TNF- α Inflammatory and Anti-Inflammatory Synchronization Affects Gingival Stem/Progenitor Cells' Regenerative Attributes. Stem Cells Int 2017; 2017:1349481. [PMID: 29250118 PMCID: PMC5700502 DOI: 10.1155/2017/1349481] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 09/26/2017] [Indexed: 01/16/2023] Open
Abstract
Cytokines play major roles in tissue destruction/repair. The present study investigates proliferative and osteogenic differentiation potentials of gingival mesenchymal stem/progenitor cells (G-MSCs), influenced by IL-1/TNF-α inflammatory/anti-inflammatory conditions. Human G-MSCs were isolated, characterized, and cultured in basic medium (control group, M1), in basic medium with IL-1β, TNF-α, and IFN-γ (inflammatory group, M2) and with IL-1ra/TNF-αi added to M2 (anti-inflammatory group, M3). MTT tests at days 1, 3, and 7 and CFU assay at day 12 were conducted. Osteogenic differentiation was analyzed by bone-specific transcription factors (RUNX2), alkaline phosphatase (ALP), type I collagen (Col-I), osteopontin (OPN), and osteonectin (ON) expression at days 1, 3, 7, and 14 and Alizarin red staining at day 14. At day 3, the control group showed the highest cell numbers. At day 7, cell numbers in inflammatory and anti-inflammatory group outnumbered the control group. At day 12, CFUs decreased in the inflammatory and anti-inflammatory groups, with altered cellular morphology. The anti-inflammatory group demonstrated elevated bone-specific transcription factors at 14 days. After 14 days of osteogenic induction, calcified nodules in the anti-inflammatory group were higher compared to control and inflammatory groups. For regeneration, initial inflammatory stimuli appear essential for G-MSCs' proliferation. With inflammatory persistence, this positive effect perishes and is followed by a short-term stimulatory one on osteogenesis. At this stage, selective anti-inflammatory intervention could boost G-MSCs' differentiation.
Collapse
|
83
|
Firmin FF, Oger F, Gheeraert C, Dubois-Chevalier J, Vercoutter-Edouart AS, Alzaid F, Mazuy C, Dehondt H, Alexandre J, Derudas B, Dhalluin Q, Ploton M, Berthier A, Woitrain E, Lefebvre T, Venteclef N, Pattou F, Staels B, Eeckhoute J, Lefebvre P. The RBM14/CoAA-interacting, long intergenic non-coding RNA Paral1 regulates adipogenesis and coactivates the nuclear receptor PPARγ. Sci Rep 2017; 7:14087. [PMID: 29075020 PMCID: PMC5658386 DOI: 10.1038/s41598-017-14570-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/12/2017] [Indexed: 02/03/2023] Open
Abstract
Adipocyte differentiation and function relies on a network of transcription factors, which is disrupted in obesity-associated low grade, chronic inflammation leading to adipose tissue dysfunction. In this context, there is a need for a thorough understanding of the transcriptional regulatory network involved in adipose tissue pathophysiology. Recent advances in the functional annotation of the genome has highlighted the role of non-coding RNAs in cellular differentiation processes in coordination with transcription factors. Using an unbiased genome-wide approach, we identified and characterized a novel long intergenic non-coding RNA (lincRNA) strongly induced during adipocyte differentiation. This lincRNA favors adipocyte differentiation and coactivates the master adipogenic regulator peroxisome proliferator-activated receptor gamma (PPARγ) through interaction with the paraspeckle component and hnRNP-like RNA binding protein 14 (RBM14/NCoAA), and was therefore called PPARγ-activator RBM14-associated lncRNA (Paral1). Paral1 expression is restricted to adipocytes and decreased in humans with increasing body mass index. A decreased expression was also observed in diet-induced or genetic mouse models of obesity and this down-regulation was mimicked in vitro by TNF treatment. In conclusion, we have identified a novel component of the adipogenic transcriptional regulatory network defining the lincRNA Paral1 as an obesity-sensitive regulator of adipocyte differentiation and function.
Collapse
Affiliation(s)
- François F Firmin
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Frederik Oger
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Céline Gheeraert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Julie Dubois-Chevalier
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Anne-Sophie Vercoutter-Edouart
- CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, FRABio FR 3688, Univ, Lille, Villeneuve d'Ascq, F-59650, France
| | - Fawaz Alzaid
- INSERM UMRS 1138, Sorbonne Universités, UPMC Université Paris 06; Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; and Centre de Recherche des Cordeliers, Paris, F-75006, France
| | - Claire Mazuy
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Hélène Dehondt
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Jeremy Alexandre
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Bruno Derudas
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Quentin Dhalluin
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Maheul Ploton
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Alexandre Berthier
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Eloise Woitrain
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Tony Lefebvre
- CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, FRABio FR 3688, Univ, Lille, Villeneuve d'Ascq, F-59650, France
| | - Nicolas Venteclef
- INSERM UMRS 1138, Sorbonne Universités, UPMC Université Paris 06; Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; and Centre de Recherche des Cordeliers, Paris, F-75006, France
| | - François Pattou
- Univ. Lille, Inserm, CHU Lille, U1190- EGID, F-59000, Lille, France
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Jérôme Eeckhoute
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Philippe Lefebvre
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France.
| |
Collapse
|
84
|
Kan C, Chen L, Hu Y, Lu H, Li Y, Kessler JA, Kan L. Microenvironmental factors that regulate mesenchymal stem cells: lessons learned from the study of heterotopic ossification. Histol Histopathol 2017; 32:977-985. [PMID: 28328009 PMCID: PMC5809774 DOI: 10.14670/hh-11-890] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bone marrow contains a non-hematopoietic, clonogenic, multipotent population of stromal cells that are later called mesenchymal stem cells (MSC). Similar cells that share many common features with MSC are also found in other organs, which are thought to contribute both to normal tissue regeneration and to pathological processes such as heterotopic ossification (HO), the formation of ectopic bone in soft tissue. Understanding the microenvironmental factors that regulate MSC in vivo is essential both for understanding the biology of the stem cells and for effective translational applications of MSC. Unfortunately, this important aspect has been largely underappreciated. This review tries to raise the attention and highlight this critical issue by updating the relevant literature along with discussions of the key issues in the area.
Collapse
Affiliation(s)
- Chen Kan
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lijun Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yangyang Hu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Haimei Lu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yuyun Li
- Department of Medical Laboratory Science, Bengbu Medical College, Bengbu, China
| | - John A Kessler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lixin Kan
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Department of Medical Laboratory Science, Bengbu Medical College, Bengbu, China
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
85
|
Zhou L, Dörfer CE, Chen L, Fawzy El-Sayed KM. Porphyromonas gingivalislipopolysaccharides affect gingival stem/progenitor cells attributes through NF-κB, but not Wnt/β-catenin, pathway. J Clin Periodontol 2017; 44:1112-1122. [DOI: 10.1111/jcpe.12777] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Lili Zhou
- Clinic of Conservative Dentistry and Periodontology; School of Dental Medicine; Christian-Albrechts Universität at Kiel; Kiel Germany
- Department of Oral Medicine; The Second Affiliated Hospital; School of Medicine; Zhejiang University; Hangzhou China
| | - Christof E. Dörfer
- Clinic of Conservative Dentistry and Periodontology; School of Dental Medicine; Christian-Albrechts Universität at Kiel; Kiel Germany
| | - Lili Chen
- Department of Oral Medicine; The Second Affiliated Hospital; School of Medicine; Zhejiang University; Hangzhou China
| | - Karim M. Fawzy El-Sayed
- Clinic of Conservative Dentistry and Periodontology; School of Dental Medicine; Christian-Albrechts Universität at Kiel; Kiel Germany
- Oral Medicine and Periodontology Department; Faculty of Oral and Dental Medicine; Cairo University; Cairo Egypt
| |
Collapse
|
86
|
Córdova LA, Loi F, Lin TH, Gibon E, Pajarinen J, Nabeshima A, Lu L, Yao Z, Goodman SB. CCL2, CCL5, and IGF-1 participate in the immunomodulation of osteogenesis during M1/M2 transition in vitro. J Biomed Mater Res A 2017; 105:3069-3076. [PMID: 28782174 DOI: 10.1002/jbm.a.36166] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 06/21/2017] [Accepted: 08/01/2017] [Indexed: 12/20/2022]
Abstract
The modulation of macrophage phenotype from pro-inflammatory (M1) to tissue healing (M2) via exogenous addition of interleukin-4 (IL-4) facilitates osteogenesis; however, the molecular mediators underlying this phenomenon remain unknown. This study characterizes the IL-4-dependent paracrine crosstalk between macrophages and osteoprogenitors and its effect on osteogenesis in vitro. Primary murine M1 were co-cultured with MC3T3 cells (M1-MC3T3) in both transwell plates and direct co-cultures. To modulate M1 to M2, M1-MC3T3 were treated with IL-4 (20 ng/mL) at day 3 after seeding (M1 + IL-4-MC3T3). Selected molecular targets were assessed at days 3 and 6 after seeding at protein and mRNA levels. Mineralization was assessed at day 21. Transwell M1 + IL-4-MC3T3 significantly enhanced the secretion of CCL2/MCP-1, IGF-1 and to a lesser degree, CCL5/RANTES at day 6. At day 3, alkaline phosphatase (Alpl) was upregulated in direct M1-MC3T3. At day 6, Smurf2 and Insulin growth factor-1 (IGF-1) were downregulated and upregulated, respectively, in direct M1 + IL-4-MC3T3. Finally, M1 + IL-4-MC3T3 increased bone matrix mineralization compared with MC3T3 cells in transwell, but this was significantly less than M1-MC3T3. Taken together, macrophage subtypes enhanced the osteogenesis in transwell setting and the transition from M1 to M2 was associated with an increase in bone anabolic factors CCL2/MCP-1, CCL5/RANTES and IGF-1 in vitro. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 3069-3076, 2017.
Collapse
Affiliation(s)
- Luis A Córdova
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305.,Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Chile, Independencia, Santiago, 8380000, Chile
| | - Florence Loi
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Tzu-Hua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Emmanuel Gibon
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305.,Biomechanics and Bone & Joint Biomaterials Laboratory, Faculty of Medicine, Paris7 University, Paris, France
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Akira Nabeshima
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Laura Lu
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, 94305.,Department of Bioengineering, Stanford University, Stanford, California, 94305
| |
Collapse
|
87
|
Long bone mesenchymal stem cells (Lb-MSCs): clinically reliable cells for osteo-diseases. Cell Tissue Bank 2017; 18:489-500. [DOI: 10.1007/s10561-017-9652-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 08/08/2017] [Indexed: 01/24/2023]
|
88
|
Abstract
PURPOSE OF REVIEW In the process of bone fracture healing, inflammation is thought to be an essential process that precedes bone formation and remodeling. We review recent studies on bone fracture healing from an osteoimmunological point of view. RECENT FINDINGS Based on previous observations that many types of immune cells infiltrate into the bone injury site and release a variety of molecules, recent studies have addressed the roles of specific immune cell subsets. Macrophages and interleukin (IL)-17-producing γδ T cells enhance bone healing, whereas CD8+ T cells impair bone repair. Additionally, IL-10-producing B cells may contribute to bone healing by suppressing excessive and/or prolonged inflammation. Although the involvement of other cells and molecules has been suggested, the precise underlying mechanisms remain elusive. Accumulating evidence has begun to reveal the deeper picture of bone fracture healing. Further studies are required for the development of novel therapeutic strategies for bone fracture.
Collapse
Affiliation(s)
- Takehito Ono
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
89
|
Zhang JR, Pang DD, Tong Q, Liu X, Su DF, Dai SM. Different Modulatory Effects of IL-17, IL-22, and IL-23 on Osteoblast Differentiation. Mediators Inflamm 2017; 2017:5950395. [PMID: 28831209 PMCID: PMC5555000 DOI: 10.1155/2017/5950395] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 06/04/2017] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES To examine the expressions of IL-17, IL-22, and IL-23 receptors in four osteoblast models and the effects of IL-17, IL-22, and IL-23 on osteoblasts. METHODS Gene expression levels of receptors, alkaline phosphatase (ALP), osteocalcin (OCN), and Runt-related transcription factor 2 (Runx-2), were evaluated by RT-PCR and real-time RT-PCR. Proliferative responses and cell cycle analysis were detected by a CCK-8 assay and flow cytometry, respectively. ALP activity and ALP mass were detected by an ALP activity assay and ALP staining, respectively. RESULTS In primary osteoblasts, only the IL-17 receptor was expressed. In C2C12, MC3T3-E1, and Saos-2 cells, the genes of IL-17, IL-22, and IL-23 receptors were not detectable. None of IL-17, IL-22, and IL-23 had an obvious effect on the proliferation of primary osteoblasts, but IL-17 exhibited an inhibitory effect on the gene expression of ALP, OCN, and Runx-2. The ALP activity and ALP mass of primary osteoblasts were downregulated by IL-17 treatment in a dose-dependent manner, and IL-17 failed to inhibit BMP-2-induced phosphorylation of Smad. CONCLUSION Primary osteoblasts constitutively express IL-17 receptors, but none of C2C12 cells, MC3T3-E1 cells, and Saos-2 cells express any receptors for IL-17, IL-22, and IL-23. IL-17 inhibits BMP-2-induced osteoblast differentiation via the BMP/Smad-independent pathway.
Collapse
Affiliation(s)
- Jing-Ru Zhang
- Department of Rheumatology & Immunology, Changhai Hospital, Second Military Medical University, Shanghai, China
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Dan-Dan Pang
- Department of Rheumatology & Immunology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qiang Tong
- Department of Rheumatology & Immunology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xia Liu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Ding-Feng Su
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Sheng-Ming Dai
- Department of Rheumatology & Immunology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
90
|
Barrachina L, Remacha AR, Romero A, Vázquez FJ, Albareda J, Prades M, Ranera B, Zaragoza P, Martín-Burriel I, Rodellar C. Inflammation affects the viability and plasticity of equine mesenchymal stem cells: possible implications in intra-articular treatments. J Vet Sci 2017; 18:39-49. [PMID: 27297420 PMCID: PMC5366301 DOI: 10.4142/jvs.2017.18.1.39] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/19/2016] [Accepted: 05/12/2016] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are gaining relevance for treating equine joint injuries because of their ability to limit inflammation and stimulate regeneration. Because inflammation activates MSC immunoregulatory function, proinflammatory priming could improve MSC efficacy. However, inflammatory molecules present in synovial fluid or added to the culture medium might have deleterious effects on MSCs. Therefore, this study was conducted to investigate the effects of inflammatory synovial fluid and proinflammatory cytokines priming on viability and plasticity of equine MSCs. Equine bone marrow derived MSCs (eBM-MSCs) from three animals were cultured for 72 h in media supplemented with: 20% inflammatory synovial fluid (SF); 50 ng/mL IFN-γ and TNF-α (CK50); and 20 ng/mL IFN-γ and TNF-α (CK20). Proliferation assay and expression of proliferation and apoptosis-related genes showed that SF exposed-eBM-MSCs maintained their viability, whereas the viability of CK primed-eBM-MSCs was significantly impaired. Tri-lineage differentiation assay revealed that exposure to inflammatory synovial fluid did not alter eBM-MSCs differentiation potential; however, eBM-MSCs primed with cytokines did not display osteogenic, adipogenic or chondrogenic phenotype. The inflammatory synovial environment is well tolerated by eBM-MSCs, whereas cytokine priming negatively affects the viability and differentiation abilities of eBM-MSCs, which might limit their in vivo efficacy.
Collapse
Affiliation(s)
- Laura Barrachina
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain.,Service of Equine Surgery and Medicine, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Ana Rosa Remacha
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Antonio Romero
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain.,Service of Equine Surgery and Medicine, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Francisco José Vázquez
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain.,Service of Equine Surgery and Medicine, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Jorge Albareda
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain.,Service of Orthopedic Surgery and Traumatology, University Clinical Hospital Lozano Blesa, 50009 Zaragoza, Spain
| | - Marta Prades
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain.,Service of Equine Surgery, Veterinary Hospital, Autonomous University of Barcelona, 08193 Barcelona, Spain
| | - Beatriz Ranera
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Pilar Zaragoza
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Inmaculada Martín-Burriel
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Clementina Rodellar
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| |
Collapse
|
91
|
Huang J, Chen L. IL-1β inhibits osteogenesis of human bone marrow-derived mesenchymal stem cells by activating FoxD3/microRNA-496 to repress wnt signaling. Genesis 2017; 55. [PMID: 28509407 DOI: 10.1002/dvg.23040] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/03/2017] [Accepted: 05/09/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Jian Huang
- Department of Orthopaedics; Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University; 67 Dongchang west Road Liaocheng Shandong 252000 People's Republic of China
| | - Liang Chen
- Department of Orthopaedics; Liaocheng People's Hospital and Liaocheng Clinical School of Taishan Medical University; 67 Dongchang west Road Liaocheng Shandong 252000 People's Republic of China
| |
Collapse
|
92
|
Sun C, Liu F, Cen S, Chen L, Wang Y, Sun H, Deng H, Hu R. Tensile strength suppresses the osteogenesis of periodontal ligament cells in inflammatory microenvironments. Mol Med Rep 2017; 16:666-672. [PMID: 28560407 PMCID: PMC5482070 DOI: 10.3892/mmr.2017.6644] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 02/15/2017] [Indexed: 12/16/2022] Open
Abstract
The present study aimed to investigate the role of orthodontic force in osteogenesis differentiation, matrix deposition and mineralization in periodontal ligament cells (PDLCs) cells in inflammatory microenvironments. The mesenchymal origin of PDLCs was confirmed by vimentin and cytokeratin staining. PDLCs were exposed to inflammatory cytokines (5 ng/ml IL‑1β and 10 ng/ml TNF‑α) and/or tensile strength (0.5 Hz, 12% elongation) for 12, 24 or 48 h. Cell proliferation and tensile strength‑induced cytokine expression were assessed by MTT assay and ELISA, respectively. Runt‑related transcription factor 2 (RUNX2) and type I collagen (COL‑I) expression were analysed by reverse transcription‑quantitative polymerase chain reaction and western blot analysis. Additionally, alkaline phosphatase activity was measured, and the mineralization profile was evaluated by alizarin red S staining. PDLCs exposed to tensile strength in inflammatory microenvironments exhibited reduced proliferation and mineralization potential. Treatment with the inflammatory cytokines IL‑1β and TNF‑α increased RUNX2 expression levels; however, decreased COL‑I expression levels, indicating that bone formation and matrix deposition involve different mechanisms in PDL tissues. Notably, RUNX2 and COL‑I expression levels were decreased in PDLCs exposed to a combination of an inflammatory environment and loading strength. The decreased osteogenic potential in an inflammatory microenvironment under tensile strength suggests that orthodontic force may amplify periodontal destruction in orthodontic patients with periodontitis.
Collapse
Affiliation(s)
- Chaofan Sun
- Department of Orthodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Fen Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Shendan Cen
- Department of Periodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Lijiao Chen
- Department of Orthodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Yi Wang
- Faculty of Dentistry, University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Hao Sun
- Department of Orthodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Hui Deng
- Department of Periodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Rongdang Hu
- Department of Orthodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| |
Collapse
|
93
|
Platas J, Guillén MI, Gomar F, Castejón MA, Esbrit P, Alcaraz MJ. Anti-senescence and Anti-inflammatory Effects of the C-terminal Moiety of PTHrP Peptides in OA Osteoblasts. J Gerontol A Biol Sci Med Sci 2017; 72:624-631. [PMID: 27271252 DOI: 10.1093/gerona/glw100] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/16/2016] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is characterized by degenerative changes in the whole joint leading to physical disability in the elderly population. This condition is associated with altered bone metabolism in subchondral areas suggesting that therapeutic strategies aimed at modifying bone cell metabolism may be of interest. We have investigated the effects of several parathyroid hormone-related protein (PTHrP)-derived peptides (1-37): (N-terminal), (107-111) and (107-139) (C-terminal) on senescence features induced by inflammatory stress in human OA osteoblasts. Incubation of these primary cells with interleukin(IL)-1β led to an increased expression of senescence markers senescence-associated-β-galactosidase activity, γH2AX foci, p16, p21, p53, and caveolin-1. PTHrP (107-111) and PTHrP (107-139) significantly reduced all these parameters. Both peptides decreased the production of IL-6 and prostaglandin E2 which was the consequence of cyclo-oxygenase-2 downregulation. PTHrP (107-139) also reduced tumor necrosis factor-α release. These anti-inflammatory effects would be related to the reduction of nuclear factor-κB activation by both peptides and activator protein-1 by PTHrP (107-139). The three PTHrP peptides favored osteoblastic function although the C-terminal domain of PTHrP was more efficient than its N-terminal domain. Our data support an anti-senescence and anti-inflammatory role for the C-terminal moiety of PTHrP with potential applications in chronic inflammatory conditions such as OA.
Collapse
Affiliation(s)
- Julia Platas
- Department of Pharmacology and IDM, University of Valencia, Valencia, Spain.,Cooperative Research Thematic Network on Aging and Frailty (RETICEF), ISCIII, Madrid, Spain
| | - Maria Isabel Guillén
- Department of Pharmacology and IDM, University of Valencia, Valencia, Spain.,Cooperative Research Thematic Network on Aging and Frailty (RETICEF), ISCIII, Madrid, Spain.,Department of Pharmacy, Cardenal Herrera-CEU University, Valencia, Spain
| | - Francisco Gomar
- Cooperative Research Thematic Network on Aging and Frailty (RETICEF), ISCIII, Madrid, Spain.,Department of Surgery, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Miguel Angel Castejón
- Department of Orthopaedic Surgery and Traumatology, De la Ribera University Hospital, Valencia, Spain
| | - Pedro Esbrit
- Cooperative Research Thematic Network on Aging and Frailty (RETICEF), ISCIII, Madrid, Spain.,Joint and Bone Research Unit, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Maria José Alcaraz
- Department of Pharmacology and IDM, University of Valencia, Valencia, Spain.,Cooperative Research Thematic Network on Aging and Frailty (RETICEF), ISCIII, Madrid, Spain
| |
Collapse
|
94
|
Synergistic attenuation of ovariectomy-induced bone loss by combined use of fish oil and 17β-oestradiol. Br J Nutr 2017; 117:479-489. [DOI: 10.1017/s0007114517000344] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
AbstractOestrogen and n-3 PUFA, especially EPA and DHA, have been reported to have beneficial effects on bone loss. Thus, the purpose of the present study was to investigate the synergistic bone-protective mechanism of combined treatments of EPA+DHA supplementation and oestrogen injection in ovariectomised rats. Rats were fed a modified American Institute of Nutrition-93G diet with 0 %, 1 % or 2 % n-3 PUFA (EPA+DHA) relative to the total energy intake for 12 weeks. Rats were surgically ovariectomised at week 8, and after a 1-week recovery period rats were injected with either 17β-oestradiol-3-benzoate (E2) or maize oil for the last 3 weeks. Combined use of n-3 PUFA and E2 synergistically increased femoral cortical bone volume, bone mineral content and the bone expression of runt-related transcription factor 2 (RUNX2), but decreased the bone expression of IL-1β. Both n-3 PUFA and E2 decreased the bone expressions of IL-7, TNF-α and PPAR-γ, and increased the bone expression of oestrogen receptor-α. n-3 PUFA in the presence of E2 and E2 alone significantly decreased the bone expressions of IL-1β and IL-6 and increased the bone expression of RUNX2. E2 significantly decreased the serum levels of bone turnover markers and the bone expression of receptor activator of NF-κB ligand, but decreased the bone expression of osteoprotegerin. The combined use of n-3 PUFA and E2 exerted synergistic bone-protective efficacy through up-regulation of RUNX2, an essential transcription factor for bone formation, as well as the suppression of bone-resorbing cytokine IL-1β.
Collapse
|
95
|
Garg P, Mazur MM, Buck AC, Wandtke ME, Liu J, Ebraheim NA. Prospective Review of Mesenchymal Stem Cells Differentiation into Osteoblasts. Orthop Surg 2017; 9:13-19. [PMID: 28276640 DOI: 10.1111/os.12304] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 09/27/2016] [Indexed: 01/02/2023] Open
Abstract
Stem cell research has been a popular topic in the past few decades. This review aims to discuss factors that help regulate, induce, and enhance mesenchymal stem cell (MSC) differentiation into osteoblasts for bone regeneration. The factors analyzed include bone morphogenic protein (BMP), transforming growth factor β (TGF-β), stromal cell-derived factor 1 (SDF-1), insulin-like growth factor type 1 (IGF-1), histone demethylase JMJD3, cyclin dependent kinase 1 (CDK1), fucoidan, Runx2 transcription factor, and TAZ transcriptional coactivator. Methods promoting bone healing are also evaluated in this review that have shown promise in previous studies. Methods tested using animal models include low intensity pulsed ultrasound (LIPUS) with MSC, micro motion, AMD3100 injections, BMP delivery, MSC transplantation, tissue engineering utilizing scaffolds, anti-IL-20 monoclonal antibody, low dose photodynamic therapy, and bone marrow stromal cell transplants. Human clinical trial methods analyzed include osteoblast injections, bone marrow grafts, bone marrow and platelet rich plasma transplantation, tissue engineering using scaffolds, and recombinant human BMP-2. These methods have been shown to promote and accelerate new bone formation. These various methods for enhanced bone regeneration have the potential to be used, following further research, in clinical practice.
Collapse
Affiliation(s)
- Priyanka Garg
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Matthew M Mazur
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Amy C Buck
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Meghan E Wandtke
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Jiayong Liu
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| | - Nabil A Ebraheim
- Department of Orthopaedic Surgery, University of Toledo Medical Center, Toledo, Ohio, USA
| |
Collapse
|
96
|
Barrachina L, Remacha AR, Romero A, Vázquez FJ, Albareda J, Prades M, Gosálvez J, Roy R, Zaragoza P, Martín-Burriel I, Rodellar C. Priming Equine Bone Marrow-Derived Mesenchymal Stem Cells with Proinflammatory Cytokines: Implications in Immunomodulation–Immunogenicity Balance, Cell Viability, and Differentiation Potential. Stem Cells Dev 2017; 26:15-24. [DOI: 10.1089/scd.2016.0209] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Laura Barrachina
- Laboratorio de Genetica Bioquimica LAGENBIO (Universidad de Zaragoza), Instituto Agroalimentario de Aragon– IA2 – (Universidad de Zaragoza-CITA), Instituto de Investigacion Sanitaria de Aragon (IIS), Zaragoza, Spain
- Servicio de Cirugía y Medicina Equina, Hospital Veterinario, Universidad de Zaragoza, Zaragoza, Spain
| | - Ana Rosa Remacha
- Laboratorio de Genetica Bioquimica LAGENBIO (Universidad de Zaragoza), Instituto Agroalimentario de Aragon– IA2 – (Universidad de Zaragoza-CITA), Instituto de Investigacion Sanitaria de Aragon (IIS), Zaragoza, Spain
| | - Antonio Romero
- Laboratorio de Genetica Bioquimica LAGENBIO (Universidad de Zaragoza), Instituto Agroalimentario de Aragon– IA2 – (Universidad de Zaragoza-CITA), Instituto de Investigacion Sanitaria de Aragon (IIS), Zaragoza, Spain
- Servicio de Cirugía y Medicina Equina, Hospital Veterinario, Universidad de Zaragoza, Zaragoza, Spain
| | - Francisco José Vázquez
- Laboratorio de Genetica Bioquimica LAGENBIO (Universidad de Zaragoza), Instituto Agroalimentario de Aragon– IA2 – (Universidad de Zaragoza-CITA), Instituto de Investigacion Sanitaria de Aragon (IIS), Zaragoza, Spain
- Servicio de Cirugía y Medicina Equina, Hospital Veterinario, Universidad de Zaragoza, Zaragoza, Spain
| | - Jorge Albareda
- Laboratorio de Genetica Bioquimica LAGENBIO (Universidad de Zaragoza), Instituto Agroalimentario de Aragon– IA2 – (Universidad de Zaragoza-CITA), Instituto de Investigacion Sanitaria de Aragon (IIS), Zaragoza, Spain
- Servicio de Cirugía Ortopédica y Traumatología, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Zaragoza, Spain
| | - Marta Prades
- Laboratorio de Genetica Bioquimica LAGENBIO (Universidad de Zaragoza), Instituto Agroalimentario de Aragon– IA2 – (Universidad de Zaragoza-CITA), Instituto de Investigacion Sanitaria de Aragon (IIS), Zaragoza, Spain
- Departament de Medicina i Cirugia Animal, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Jaime Gosálvez
- Departamento de Biología, Universidad Autónoma de Madrid, Ciudad Universitaria de Cantoblanco, Madrid, Spain
| | - Rosa Roy
- Departamento de Biología, Universidad Autónoma de Madrid, Ciudad Universitaria de Cantoblanco, Madrid, Spain
| | - Pilar Zaragoza
- Laboratorio de Genetica Bioquimica LAGENBIO (Universidad de Zaragoza), Instituto Agroalimentario de Aragon– IA2 – (Universidad de Zaragoza-CITA), Instituto de Investigacion Sanitaria de Aragon (IIS), Zaragoza, Spain
| | - Inmaculada Martín-Burriel
- Laboratorio de Genetica Bioquimica LAGENBIO (Universidad de Zaragoza), Instituto Agroalimentario de Aragon– IA2 – (Universidad de Zaragoza-CITA), Instituto de Investigacion Sanitaria de Aragon (IIS), Zaragoza, Spain
| | - Clementina Rodellar
- Laboratorio de Genetica Bioquimica LAGENBIO (Universidad de Zaragoza), Instituto Agroalimentario de Aragon– IA2 – (Universidad de Zaragoza-CITA), Instituto de Investigacion Sanitaria de Aragon (IIS), Zaragoza, Spain
| |
Collapse
|
97
|
Reckenbeil J, Kraus D, Stark H, Rath-Deschner B, Jäger A, Wenghoefer M, Winter J, Götz W. Insulin-like growth factor 1 (IGF1) affects proliferation and differentiation and wound healing processes in an inflammatory environment with p38 controlling early osteoblast differentiation in periodontal ligament cells. Arch Oral Biol 2017; 73:142-150. [DOI: 10.1016/j.archoralbio.2016.10.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 08/31/2016] [Accepted: 10/11/2016] [Indexed: 12/21/2022]
|
98
|
Lin TH, Pajarinen J, Lu L, Nabeshima A, Cordova LA, Yao Z, Goodman SB. NF-κB as a Therapeutic Target in Inflammatory-Associated Bone Diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 107:117-154. [PMID: 28215222 DOI: 10.1016/bs.apcsb.2016.11.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Inflammation is a defensive mechanism for pathogen clearance and maintaining tissue homeostasis. In the skeletal system, inflammation is closely associated with many bone disorders including fractures, nonunions, periprosthetic osteolysis (bone loss around orthopedic implants), and osteoporosis. Acute inflammation is a critical step for proper bone-healing and bone-remodeling processes. On the other hand, chronic inflammation with excessive proinflammatory cytokines disrupts the balance of skeletal homeostasis involving osteoblastic (bone formation) and osteoclastic (bone resorption) activities. NF-κB is a transcriptional factor that regulates the inflammatory response and bone-remodeling processes in both bone-forming and bone-resorption cells. In vitro and in vivo evidences suggest that NF-κB is an important potential therapeutic target for inflammation-associated bone disorders by modulating inflammation and bone-remodeling process simultaneously. The challenges of NF-κB-targeting therapy in bone disorders include: (1) the complexity of canonical and noncanonical NF-κB pathways; (2) the fundamental roles of NF-κB-mediated signaling for bone regeneration at earlier phases of tissue damage and acute inflammation; and (3) the potential toxic effects on nontargeted cells such as lymphocytes. Recent developments of novel inhibitors with differential approaches to modulate NF-κB activity, and the controlled release (local) or bone-targeting drug delivery (systemic) strategies, have largely increased the translational application of NF-κB therapy in bone disorders. Taken together, temporal modulation of NF-κB pathways with the combination of recent advanced bone-targeting drug delivery techniques is a highly translational strategy to reestablish homeostasis in the skeletal system.
Collapse
Affiliation(s)
- T-H Lin
- Stanford University, Stanford, CA, United States
| | - J Pajarinen
- Stanford University, Stanford, CA, United States
| | - L Lu
- Stanford University, Stanford, CA, United States
| | - A Nabeshima
- Stanford University, Stanford, CA, United States
| | - L A Cordova
- Stanford University, Stanford, CA, United States; Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Z Yao
- Stanford University, Stanford, CA, United States
| | - S B Goodman
- Stanford University, Stanford, CA, United States.
| |
Collapse
|
99
|
Fakhry M, Roszkowska M, Briolay A, Bougault C, Guignandon A, Diaz-Hernandez JI, Diaz-Hernandez M, Pikula S, Buchet R, Hamade E, Badran B, Bessueille L, Magne D. TNAP stimulates vascular smooth muscle cell trans-differentiation into chondrocytes through calcium deposition and BMP-2 activation: Possible implication in atherosclerotic plaque stability. Biochim Biophys Acta Mol Basis Dis 2016; 1863:643-653. [PMID: 27932058 DOI: 10.1016/j.bbadis.2016.12.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/12/2016] [Accepted: 12/04/2016] [Indexed: 01/15/2023]
Abstract
Atherosclerotic plaque calcification varies from early, diffuse microcalcifications to a bone-like tissue formed by endochondral ossification. Recently, a paradigm has emerged suggesting that if the bone metaplasia stabilizes the plaques, microcalcifications are harmful. Tissue-nonspecific alkaline phosphatase (TNAP), an ectoenzyme necessary for mineralization by its ability to hydrolyze inorganic pyrophosphate (PPi), is stimulated by inflammation in vascular smooth muscle cells (VSMCs). Our objective was to determine the role of TNAP in trans-differentiation of VSMCs and calcification. In rodent MOVAS and A7R5 VSMCs, addition of exogenous alkaline phosphatase (AP) or TNAP overexpression was sufficient to stimulate the expression of several chondrocyte markers and induce mineralization. Addition of exogenous AP to human mesenchymal stem cells cultured in pellets also stimulated chondrogenesis. Moreover, TNAP inhibition with levamisole in mouse primary chondrocytes dropped mineralization as well as the expression of chondrocyte markers. VSMCs trans-differentiated into chondrocyte-like cells, as well as primary chondrocytes, used TNAP to hydrolyze PPi, and PPi provoked the same effects as TNAP inhibition in primary chondrocytes. Interestingly, apatite crystals, associated or not to collagen, mimicked the effects of TNAP on VSMC trans-differentiation. AP and apatite crystals increased the expression of BMP-2 in VSMCs, and TNAP inhibition reduced BMP-2 levels in chondrocytes. Finally, the BMP-2 inhibitor noggin blocked the rise in aggrecan induced by AP in VSMCs, suggesting that TNAP induction in VSMCs triggers calcification, which stimulates chondrogenesis through BMP-2. Endochondral ossification in atherosclerotic plaques may therefore be induced by crystals, probably to confer stability to plaques with microcalcifications.
Collapse
Affiliation(s)
- Maya Fakhry
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France; Lebanese University, Laboratory of Cancer Biology and Molecular Immunology, EDST-PRASE, Hadath-Beirut, Lebanon
| | - Monika Roszkowska
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France; Laboratory of Biochemistry of Lipids, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Anne Briolay
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - Carole Bougault
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - Alain Guignandon
- Univ Lyon, Université Jean Monnet Saint-Etienne, LBTO, UMR INSERM 1059, F-42023 Saint-Etienne, France
| | - Juan Ignacio Diaz-Hernandez
- Universidad Complutense de Madrid, Facultad de Veterinaria, Dpt. Bioquimica y Biologia Molecular IV, Madrid, Spain
| | - Miguel Diaz-Hernandez
- Universidad Complutense de Madrid, Facultad de Veterinaria, Dpt. Bioquimica y Biologia Molecular IV, Madrid, Spain
| | - Slawomir Pikula
- Laboratory of Biochemistry of Lipids, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - René Buchet
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - Eva Hamade
- Lebanese University, Laboratory of Cancer Biology and Molecular Immunology, EDST-PRASE, Hadath-Beirut, Lebanon
| | - Bassam Badran
- Lebanese University, Laboratory of Cancer Biology and Molecular Immunology, EDST-PRASE, Hadath-Beirut, Lebanon
| | | | - David Magne
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France.
| |
Collapse
|
100
|
|