1
|
Xu Q, Luo Y, Chao Z, Zhang J, Liu X, Tang Q, Wang K, Tan S, Fang M. Integrated Analysis of Transcriptome Expression Profiles Reveals miRNA-326-NKX3.2-Regulated Porcine Chondrocyte Differentiation. Int J Mol Sci 2023; 24:ijms24087257. [PMID: 37108419 PMCID: PMC10138716 DOI: 10.3390/ijms24087257] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
The porcine body length trait is an essential factor affecting meat production and reproductive performance. It is evident that the development/lengthening of individual vertebrae is one of the main reasons for increases in body length; however, the underlying molecular mechanism remains unclear. In this study, RNA-seq analysis was used to profile the transcriptome (lncRNA, mRNA, and miRNA) of the thoracic intervertebral cartilage (TIC) at two time points (1 and 4 months) during vertebral column development in Yorkshire (Y) and Wuzhishan pigs (W). There were four groups: 1- (Y1) and 4-month-old (Y4) Yorkshire pigs and 1- (W1) and 4-month-old (W4) Wuzhishan pigs. In total, 161, 275, 86, and 126 differentially expressed (DE) lncRNAs, 1478, 2643, 404, and 750 DE genes (DEGs), and 74,51, 34, and 23 DE miRNAs (DE miRNAs) were identified in the Y4 vs. Y1, W4 vs. W1, Y4 vs. W4, and Y1 vs. W1 comparisons, respectively. Functional analysis of these DE transcripts (DETs) demonstrated that they had participated in various biological processes, such as cellular component organization or biogenesis, the developmental process, the metabolic process, bone development, and cartilage development. The crucial bone development-related candidate genes NK3 Homeobox 2 (NKX3.2), Wnt ligand secretion mediator (WLS), gremlin 1 (GREM1), fibroblast growth factor receptor 3 (FGFR3), hematopoietically expressed homeobox (HHEX), (collagen type XI alpha 1 chain (COL11A1), and Wnt Family Member 16 (WNT16)) were further identified by functional analysis. Moreover, lncRNA, miRNA, and gene interaction networks were constructed; a total of 55 lncRNAs, 6 miRNAs, and 7 genes formed lncRNA-gene, miRNA-gene, and lncRNA-miRNA-gene pairs, respectively. The aim was to demonstrate that coding and non-coding genes may co-regulate porcine spine development through interaction networks. NKX3.2 was identified as being specifically expressed in cartilage tissues, and it delayed chondrocyte differentiation. miRNA-326 regulated chondrocyte differentiation by targeting NKX3.2. The present study provides the first non-coding RNA and gene expression profiles in the porcine TIC, constructs the lncRNA-miRNA-gene interaction networks, and confirms the function of NKX3.2 in vertebral column development. These findings contribute to the understanding of the potential molecular mechanisms regulating pig vertebral column development. They expand our knowledge about the differences in body length between different pig species and provide a foundation for future studies.
Collapse
Affiliation(s)
- Qiao Xu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yabiao Luo
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhe Chao
- Institute of Animal Sciences and Veterinary, Hainan Academy of Agricultural Sciences, Haikou 571100, China
| | - Jibin Zhang
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA 91006, USA
| | - Ximing Liu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Qiguo Tang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Kejun Wang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shuyi Tan
- Institute of Animal Sciences and Veterinary, Hainan Academy of Agricultural Sciences, Haikou 571100, China
| | - Meiying Fang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
2
|
Murugaiyan K, Amirthalingam S, Hwang NSY, Jayakumar R. Role of FGF-18 in Bone Regeneration. J Funct Biomater 2023; 14:jfb14010036. [PMID: 36662083 PMCID: PMC9864085 DOI: 10.3390/jfb14010036] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
In tissue engineering, three key components are cells, biological/mechanical cues, and scaffolds. Biological cues are normally proteins such as growth factors and their derivatives, bioactive molecules, and the regulators of a gene. Numerous growth factors such as VEGF, FGF, and TGF-β are being studied and applied in different studies. The carriers used to release these growth factors also play an important role in their functioning. From the early part of the 1990s, more research has beenconductedon the role of fibroblast growth factors on the various physiological functions in our body. The fibroblast growth factor family contains 22 members. Fibroblast growth factors such as 2, 9, and 18 are mainly associated with the differentiation of osteoblasts and in bone regeneration. FGF-18 stimulates the PI3K/ERK pathway and smad1/5/8 pathway mediated via BMP-2 by blocking its antagonist, which is essential for bone formation. FGF-18 incorporated hydrogel and scaffolds had showed enhanced bone regeneration. This review highlights these functions and current trends using this growth factor and potential outcomes in the field of bone regeneration.
Collapse
Affiliation(s)
- Kavipriya Murugaiyan
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | | | - Nathaniel Suk-Yeon Hwang
- Institute of Engineering Research, Seoul National University, Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- BioMAX/N-Bio, Institute of BioEngineering, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Rangasamy Jayakumar
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India
- Correspondence: or
| |
Collapse
|
3
|
Hoshi M, Taira M, Sawada T, Hachinohe Y, Hatakeyama W, Takafuji K, Tekemoto S, Kondo H. Preparation of Collagen/Hydroxyapatite Composites Using the Alternate Immersion Method and Evaluation of the Cranial Bone-Forming Capability of Composites Complexed with Acidic Gelatin and b-FGF. MATERIALS (BASEL, SWITZERLAND) 2022; 15:8802. [PMID: 36556608 PMCID: PMC9787395 DOI: 10.3390/ma15248802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 06/17/2023]
Abstract
Bone-substitute materials are essential in dental implantology. We prepared collagen (Col)/hydroxyapatite (Hap)/acidic gelatin (AG)/basic fibroblast growth factor (b-FGF) constructs with enhanced bone-forming capability. The Col/Hap apatite composites were prepared by immersing Col sponges alternately in calcium and phosphate ion solutions five times, for 20 and 60 min, respectively. Then, the sponges were heated to 56 °C for 48 h. Scanning electron microscopy/energy-dispersive X-ray spectroscopy, Fourier-transform infrared spectroscopy, and X-ray diffraction analyses showed that the Col/Hap composites contained poorly crystalline Hap precipitates on the Col matrix. Col/Hap composite granules were infiltrated by AG, freeze-dried, and immersed in b-FGF solution. The wet quaternary constructs were implanted in rat cranial bone defects for 8 weeks, followed by soft X-ray measurements and histological analysis. Animal studies have shown that the constructs moderately increase bone formation in cranial bone defects. We found that an alternate immersion time of 20 min led to the greatest bone formation (p < 0.05). Constructs placed inside defects slightly extend the preexisting bone from the defect edges and lead to the formation of small island-like bones inside the defect, followed by disappearance of the constructs. The combined use of Col, Hap, AG, and b-FGF might bring about novel bone-forming biomaterials.
Collapse
Affiliation(s)
- Miki Hoshi
- Department of Prosthodontics and Oral Implantology, School of Dentistry, Iwate Medical University, 19-1 Uchimaru, Morioka 020-8505, Japan
| | - Masayuki Taira
- Department of Biomedical Engineering, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho 028-3694, Japan
| | - Tomofumi Sawada
- Department of Biomedical Engineering, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho 028-3694, Japan
| | - Yuki Hachinohe
- Department of Prosthodontics and Oral Implantology, School of Dentistry, Iwate Medical University, 19-1 Uchimaru, Morioka 020-8505, Japan
| | - Wataru Hatakeyama
- Department of Prosthodontics and Oral Implantology, School of Dentistry, Iwate Medical University, 19-1 Uchimaru, Morioka 020-8505, Japan
| | - Kyoko Takafuji
- Department of Prosthodontics and Oral Implantology, School of Dentistry, Iwate Medical University, 19-1 Uchimaru, Morioka 020-8505, Japan
| | - Shinji Tekemoto
- Department of Biomedical Engineering, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho 028-3694, Japan
| | - Hisatomo Kondo
- Department of Prosthodontics and Oral Implantology, School of Dentistry, Iwate Medical University, 19-1 Uchimaru, Morioka 020-8505, Japan
| |
Collapse
|
4
|
Andreev D, Kachler K, Schett G, Bozec A. Rheumatoid arthritis and osteoimmunology: The adverse impact of a deregulated immune system on bone metabolism. Bone 2022; 162:116468. [PMID: 35688359 DOI: 10.1016/j.bone.2022.116468] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 11/20/2022]
Abstract
The term osteoimmunology describes an interdisciplinary research field that links the investigation of osteology (bone cells) with immunology. The crosstalk between innate and adaptive immune cells and cells involved in bone remodeling, mainly bone-resorbing osteoclasts and bone-forming osteoblasts, becomes particularly obvious in the inflammatory autoimmune disease rheumatoid arthritis (RA). Besides striking inflammation of the joints, RA causes bone loss, leading to joint damage and disabilities as well as generalized osteoporosis. Mechanistically, RA-associated immune cells (macrophages, T cells, B cells etc.) produce high levels of pro-inflammatory cytokines, receptor activator of nuclear factor κB ligand (RANKL) and autoantibodies that promote bone degradation and at the same time counteract new bone formation. Today, antirheumatic therapy effectively ceases joint inflammation and arrests bone erosion. However, the repair of established bone lesions still presents a challenging task and requires improved treatment options. In this review, we outline the knowledge gained over the past years about the immunopathogenesis of RA and the impact of a dysregulated immune system on bone metabolism.
Collapse
Affiliation(s)
- Darja Andreev
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Katerina Kachler
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany.
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany.
| |
Collapse
|
5
|
Wawrzyniak A, Balawender K. Structural and Metabolic Changes in Bone. Animals (Basel) 2022; 12:ani12151946. [PMID: 35953935 PMCID: PMC9367262 DOI: 10.3390/ani12151946] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/24/2022] [Accepted: 07/27/2022] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Bone is an extremely metabolically active tissue that is regenerated and repaired over its lifetime by bone remodeling. Most bone diseases are caused by abnormal restructure processes that undermine bone structure and mechanical strength and trigger clinical symptoms, such as pain, deformity, fracture, and abnormalities of calcium and phosphate homoeostasis. The article examines the main aspects of bone development, anatomy, structure, and the mechanisms of cell and molecular regulation of bone remodeling. Abstract As an essential component of the skeleton, bone tissue provides solid support for the body and protects vital organs. Bone tissue is a reservoir of calcium, phosphate, and other ions that can be released or stored in a controlled manner to provide constant concentration in body fluids. Normally, bone development or osteogenesis occurs through two ossification processes (intra-articular and intra-chondral), but the first produces woven bone, which is quickly replaced by stronger lamellar bone. Contrary to commonly held misconceptions, bone is a relatively dynamic organ that undergoes significant turnover compared to other organs in the body. Bone metabolism is a dynamic process that involves simultaneous bone formation and resorption, controlled by numerous factors. Bone metabolism comprises the key actions. Skeletal mass, structure, and quality are accrued and maintained throughout life, and the anabolic and catabolic actions are mostly balanced due to the tight regulation of the activity of osteoblasts and osteoclasts. This activity is also provided by circulating hormones and cytokines. Bone tissue remodeling processes are regulated by various biologically active substances secreted by bone tissue cells, namely RANK, RANKL, MMP-1, MMP-9, or type 1 collagen. Bone-derived factors (BDF) influence bone function and metabolism, and pathophysiological conditions lead to bone dysfunction. This work aims to analyze and evaluate the current literature on various local and systemic factors or immune system interactions that can affect bone metabolism and its impairments.
Collapse
|
6
|
You J, Zhang Y, Zhou Y. Strontium Functionalized in Biomaterials for Bone Tissue Engineering: A Prominent Role in Osteoimmunomodulation. Front Bioeng Biotechnol 2022; 10:928799. [PMID: 35875505 PMCID: PMC9298737 DOI: 10.3389/fbioe.2022.928799] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/13/2022] [Indexed: 12/24/2022] Open
Abstract
With the development of bone tissue engineering bio-scaffold materials by adding metallic ions to improve bone healing have been extensively explored in the past decades. Strontium a non-radioactive element, as an essential osteophilic trace element for the human body, has received widespread attention in the medical field due to its superior biological properties of inhibiting bone resorption and promoting osteogenesis. As the concept of osteoimmunology developed, the design of orthopedic biomaterials has gradually shifted from “immune-friendly” to “immunomodulatory” with the aim of promoting bone healing by modulating the immune microenvironment through implanted biomaterials. The process of bone healing can be regarded as an immune-induced procedure in which immune cells can target the effector cells such as macrophages, neutrophils, osteocytes, and osteoprogenitor cells through paracrine mechanisms, affecting pathological alveolar bone resorption and physiological bone regeneration. As a kind of crucial immune cell, macrophages play a critical role in the early period of wound repair and host defense after biomaterial implantation. Despite Sr-doped biomaterials being increasingly investigated, how extracellular Sr2+ guides the organism toward favorable osteogenesis by modulating macrophages in the bone tissue microenvironment has rarely been studied. This review focuses on recent knowledge that the trace element Sr regulates bone regeneration mechanisms through the regulation of macrophage polarization, which is significant for the future development of Sr-doped bone repair materials. We will also summarize the primary mechanism of Sr2+ in bone, including calcium-sensing receptor (CaSR) and osteogenesis-related signaling pathways.
Collapse
Affiliation(s)
- Jiaqian You
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yidi Zhang
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yanmin Zhou
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
7
|
Cerqueni G, Scalzone A, Licini C, Gentile P, Mattioli-Belmonte M. Insights into oxidative stress in bone tissue and novel challenges for biomaterials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 130:112433. [PMID: 34702518 DOI: 10.1016/j.msec.2021.112433] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/28/2022]
Abstract
The presence of Reactive Oxygen Species (ROS) in bone can influence resident cells behaviour as well as the extra-cellular matrix composition and the tissue architecture. Aging, in addition to excessive overloads, unbalanced diet, smoking, predisposing genetic factors, lead to an increase of ROS and, if it is accompanied with an inappropriate production of scavengers, promotes the generation of oxidative stress that encourages bone catabolism. Furthermore, bone injuries can be triggered by numerous events such as road and sports accidents or tumour resection. Although bone tissue possesses a well-known repair and regeneration capacity, these mechanisms are inefficient in repairing large size defects and bone grafts are often necessary. ROS play a fundamental role in response after the implant introduction and can influence its success. This review provides insights on the mechanisms of oxidative stress generated by an implant in vivo and suitable ways for its modulation. The local delivery of active molecules, such as polyphenols, enhanced bone biomaterial integration evidencing that the management of the oxidative stress is a target for the effectiveness of an implant. Polyphenols have been widely used in medicine for cardiovascular, neurodegenerative, bone disorders and cancer, thanks to their antioxidant and anti-inflammatory properties. In addition, the perspective of new smart biomaterials and molecular medicine for the oxidative stress modulation in a programmable way, by the use of ROS responsive materials or by the targeting of selective molecular pathways involved in ROS generation, will be analysed and discussed critically.
Collapse
Affiliation(s)
- Giorgia Cerqueni
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60126, Italy
| | - Annachiara Scalzone
- School of Engineering, Newcastle University, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Caterina Licini
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60126, Italy; Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 204, 10129 Torino, Italy
| | - Piergiorgio Gentile
- School of Engineering, Newcastle University, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60126, Italy.
| |
Collapse
|
8
|
Myeloproliferative Disorders and its Effect on Bone Homeostasis: The Role of Megakaryocytes. Blood 2021; 139:3127-3137. [PMID: 34428274 DOI: 10.1182/blood.2021011480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/11/2021] [Indexed: 11/20/2022] Open
Abstract
Myeloproliferative Neoplasms (MPNs) are a heterogeneous group of chronic hematological diseases that arise from the clonal expansion of abnormal hematopoietic stem cells, of which Polycythemia Vera (PV), Essential Thrombocythemia (ET), and Primary Myelofibrosis (PMF) have been extensively reviewed in context of clonal expansion, fibrosis and other phenotypes. Here, we review current knowledge on the influence of different forms of MPN on bone health. Studies implicated various degrees of effect of different forms of MPN on bone density, and on osteoblast proliferation and differentiation, using murine models and human data. The majority of studies show that bone volume is generally increased in PMF patients, whereas it is slightly decreased or not altered in ET and PV patients, although possible differences between male and female phenotypes were not fully explored in most MPN forms. Osteosclerosis seen in PMF patients is a serious complication that can lead to bone marrow failure, and the loss of bone reported in some ET and PV patients can lead to osteoporotic fractures. Some MPN forms are associated with increased number of megakaryocytes (MKs), and several of the MK-associated factors in MPN are known to affect bone development. Here, we review known mechanisms involved in these processes, with focus on the role of MKs and secreted factors. Understanding MPN-associated changes in bone health could improve early intervention and treatment of this side effect of the pathology.
Collapse
|
9
|
Wazzani R, Pallu S, Bourzac C, Ahmaïdi S, Portier H, Jaffré C. Physical Activity and Bone Vascularization: A Way to Explore in Bone Repair Context? Life (Basel) 2021; 11:life11080783. [PMID: 34440527 PMCID: PMC8399402 DOI: 10.3390/life11080783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/11/2021] [Accepted: 07/21/2021] [Indexed: 01/15/2023] Open
Abstract
Physical activity is widely recognized as a biotherapy by WHO in the fight and prevention of bone diseases such as osteoporosis. It reduces the risk of disabling fractures associated with many comorbidities, and whose repair is a major public health and economic issue. Bone tissue is a dynamic supportive tissue that reshapes itself according to the mechanical stresses to which it is exposed. Physical exercise is recognized as a key factor for bone health. However, the effects of exercise on bone quality depend on exercise protocols, duration, intensity, and frequency. Today, the effects of different exercise modalities on capillary bone vascularization, bone blood flow, and bone angiogenesis remain poorly understood and unclear. As vascularization is an integral part of bone repair process, the analysis of the preventive and/or curative effects of physical exercise is currently very undeveloped. Angiogenesis–osteogenesis coupling may constitute a new way for understanding the role of physical activity, especially in fracturing or in the integration of bone biomaterials. Thus, this review aimed to clarify the link between physical activities, vascularization, and bone repair.
Collapse
Affiliation(s)
- Rkia Wazzani
- Laboratoire APERE, Université de Picardie Jules Verne, CEDEX, F-80000 Amiens, France; (R.W.); (S.A.)
| | - Stéphane Pallu
- Laboratoire B3OA, Université de Paris, CEDEX, F-75010 Paris, France; (S.P.); (C.B.); (H.P.)
- UFR Science & Technique, Université d’Orléans, CEDEX, F-45100 Orléans, France
| | - Céline Bourzac
- Laboratoire B3OA, Université de Paris, CEDEX, F-75010 Paris, France; (S.P.); (C.B.); (H.P.)
| | - Saïd Ahmaïdi
- Laboratoire APERE, Université de Picardie Jules Verne, CEDEX, F-80000 Amiens, France; (R.W.); (S.A.)
| | - Hugues Portier
- Laboratoire B3OA, Université de Paris, CEDEX, F-75010 Paris, France; (S.P.); (C.B.); (H.P.)
- UFR Science & Technique, Université d’Orléans, CEDEX, F-45100 Orléans, France
| | - Christelle Jaffré
- Laboratoire APERE, Université de Picardie Jules Verne, CEDEX, F-80000 Amiens, France; (R.W.); (S.A.)
- Laboratoire B3OA, Université de Paris, CEDEX, F-75010 Paris, France; (S.P.); (C.B.); (H.P.)
- Correspondence:
| |
Collapse
|
10
|
Vojtová L, Pavliňáková V, Muchová J, Kacvinská K, Brtníková J, Knoz M, Lipový B, Faldyna M, Göpfert E, Holoubek J, Pavlovský Z, Vícenová M, Blahnová VH, Hearnden V, Filová E. Healing and Angiogenic Properties of Collagen/Chitosan Scaffolds Enriched with Hyperstable FGF2-STAB ® Protein: In Vitro, Ex Ovo and In Vivo Comprehensive Evaluation. Biomedicines 2021; 9:590. [PMID: 34067330 PMCID: PMC8224647 DOI: 10.3390/biomedicines9060590] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/13/2021] [Accepted: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Wound healing is a process regulated by a complex interaction of multiple growth factors including fibroblast growth factor 2 (FGF2). Although FGF2 appears in several tissue engineered studies, its applications are limited due to its low stability both in vitro and in vivo. Here, this shortcoming is overcome by a unique nine-point mutant of the low molecular weight isoform FGF2 retaining full biological activity even after twenty days at 37 °C. Crosslinked freeze-dried 3D porous collagen/chitosan scaffolds enriched with this hyper stable recombinant human protein named FGF2-STAB® were tested for in vitro biocompatibility and cytotoxicity using murine 3T3-A31 fibroblasts, for angiogenic potential using an ex ovo chick chorioallantoic membrane assay and for wound healing in vivo with 3-month old white New Zealand rabbits. Metabolic activity assays indicated the positive effect of FGF2-STAB® already at very low concentrations (0.01 µg/mL). The angiogenic properties examined ex ovo showed enhanced vascularization of the tested scaffolds. Histological evaluation and gene expression analysis by RT-qPCR proved newly formed granulation tissue at the place of a previous skin defect without significant inflammation infiltration in vivo. This work highlights the safety and biocompatibility of newly developed crosslinked collagen/chitosan scaffolds involving FGF2-STAB® protein. Moreover, these sponges could be used as scaffolds for growing cells for dermis replacement, where neovascularization is a crucial parameter for successful skin regeneration.
Collapse
Affiliation(s)
- Lucy Vojtová
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
| | - Veronika Pavliňáková
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
| | - Johana Muchová
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
| | - Katarína Kacvinská
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
| | - Jana Brtníková
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
| | - Martin Knoz
- Faculty of Medicine, Department of Burns and Plastic Surgery, Institution Shared with the University Hospital Brno, 625 00 Brno, Czech Republic; (M.K.); (J.H.)
- Clinic of Plastic and Esthetic Surgery, St Anne’s University Hospital, 602 00 Brno, Czech Republic
| | - Břetislav Lipový
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
- Faculty of Medicine, Department of Burns and Plastic Surgery, Institution Shared with the University Hospital Brno, 625 00 Brno, Czech Republic; (M.K.); (J.H.)
| | - Martin Faldyna
- Veterinary Research Institute, 621 00 Brno, Czech Republic; (M.F.); (E.G.); (M.V.)
| | - Eduard Göpfert
- Veterinary Research Institute, 621 00 Brno, Czech Republic; (M.F.); (E.G.); (M.V.)
| | - Jakub Holoubek
- Faculty of Medicine, Department of Burns and Plastic Surgery, Institution Shared with the University Hospital Brno, 625 00 Brno, Czech Republic; (M.K.); (J.H.)
| | - Zdeněk Pavlovský
- Faculty of Medicine, Institute of Pathology, University Hospital Brno, Masaryk University, 625 00 Brno, Czech Republic;
| | - Monika Vícenová
- Veterinary Research Institute, 621 00 Brno, Czech Republic; (M.F.); (E.G.); (M.V.)
| | - Veronika Hefka Blahnová
- Institute of Experimental Medicine of the Czech Academy of Science, 142 20 Prague, Czech Republic; (V.H.B.); (E.F.)
| | - Vanessa Hearnden
- Department of Materials Science and Engineering, Kroto Research Institute, North Campus, University of Sheffield, Broad Lane, Sheffield S3 7HQ, UK;
| | - Eva Filová
- Institute of Experimental Medicine of the Czech Academy of Science, 142 20 Prague, Czech Republic; (V.H.B.); (E.F.)
| |
Collapse
|
11
|
Assessment of the Tissue Response to Modification of the Surface of Dental Implants with Carboxyethylphosphonic Acid and Basic Fibroblastic Growth Factor Immobilization (Fgf-2): An Experimental Study on Minipigs. BIOLOGY 2021; 10:biology10050358. [PMID: 33922433 PMCID: PMC8146140 DOI: 10.3390/biology10050358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 11/23/2022]
Abstract
Simple Summary This study aimed to evaluate the efficacy of treating the surface of dental implants with carboxyethylphosphonic acid for the immobilization of FGF-2, the influence of FGF-2 on cortical bone in close contact with dental implants, new bone formation around dental implants in the presence of FGF-2 and the influence of FGF-2 on the interthread bone area of dental implants during the healing period after insertion. Abstract The aim of this study was to evaluate the effect of implant surface treatment with carboxyethylphosphonic acid and fibroblast growth factor 2 on the bone–implant interface during the osseointegration period in vivo using an animal model. The present research was carried out in six minipigs, in whose left tibia implants were inserted as follows: eight implants with a standard surface treatment, for the control group, and eight implants with a surface treatment of carboxyethylphosphonic acid and immobilization of FGF-2, for the test group. At 4 weeks after the insertion of the implants, the animals were sacrificed for the histomorphometric analysis of the samples. The means of the results for the implant–bone contact variable (BIC) were 46.39 ± 17.49% for the test group and 34.00 ± 9.92% for the control group; the difference was not statistically significant. For the corrected implant–bone contact variable (BICc), the mean value of the test group was 60.48 ± 18.11%, and that for the control group, 43.08 ± 10.77%; the difference was statistically significant (p-value = 0.035). The new bone formation (BV/TV) showed average results of 27.28 ± 3.88% for the test group and 26.63 ± 7.90% for the control group, meaning that the differences were not statistically significant (p-value = 0.839). Regarding the bone density at the interthread level (BAI/TA), the mean value of the test group was 32.27 ± 6.70%, and that of the control group was 32.91 ± 7.76%, with a p-value of 0.863, while for the peri-implant density (BAP/TA), the mean value of the test group was 44.96 ± 7.55%, and that for the control group was 44.80 ± 8.68%, without a significant difference between the groups. The current research only found a significant difference for the bone–implant contact at the cortical level; therefore, it could be considered that FGF-2 acts on the mineralization of bone tissue. The application of carboxyethylphosphonic acid on the surface of implants can be considered a promising alternative as a biomimetic coating for the immobilization of FGF-2. Despite no differences in the new bone formation around the implants or in the interthread or peri-implant bone density being detected, the biofunctionalization of the implant surface with FGF-2 accelerates the mineralization of the bone–implant interface at the cortical level, thereby reducing the osseointegration period.
Collapse
|
12
|
Kligman S, Ren Z, Chung CH, Perillo MA, Chang YC, Koo H, Zheng Z, Li C. The Impact of Dental Implant Surface Modifications on Osseointegration and Biofilm Formation. J Clin Med 2021; 10:1641. [PMID: 33921531 PMCID: PMC8070594 DOI: 10.3390/jcm10081641] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/26/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Implant surface design has evolved to meet oral rehabilitation challenges in both healthy and compromised bone. For example, to conquer the most common dental implant-related complications, peri-implantitis, and subsequent implant loss, implant surfaces have been modified to introduce desired properties to a dental implant and thus increase the implant success rate and expand their indications. Until now, a diversity of implant surface modifications, including different physical, chemical, and biological techniques, have been applied to a broad range of materials, such as titanium, zirconia, and polyether ether ketone, to achieve these goals. Ideal modifications enhance the interaction between the implant's surface and its surrounding bone which will facilitate osseointegration while minimizing the bacterial colonization to reduce the risk of biofilm formation. This review article aims to comprehensively discuss currently available implant surface modifications commonly used in implantology in terms of their impact on osseointegration and biofilm formation, which is critical for clinicians to choose the most suitable materials to improve the success and survival of implantation.
Collapse
Affiliation(s)
- Stefanie Kligman
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Zhi Ren
- Biofilm Research Laboratories, Department of Orthodontics, Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (Z.R.); (H.K.)
| | - Chun-Hsi Chung
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (C.-H.C.); (M.A.P.)
| | - Michael Angelo Perillo
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (C.-H.C.); (M.A.P.)
| | - Yu-Cheng Chang
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Hyun Koo
- Biofilm Research Laboratories, Department of Orthodontics, Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (Z.R.); (H.K.)
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhong Zheng
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chenshuang Li
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (C.-H.C.); (M.A.P.)
| |
Collapse
|
13
|
Chang M, Lin H, Fu H, Wang J, Yang Y, Wan Z, Han G. CREB activation affects mesenchymal stem cell migration and differentiation in periodontal tissues due to orthodontic force. Int J Biochem Cell Biol 2020; 129:105862. [PMID: 33045372 DOI: 10.1016/j.biocel.2020.105862] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/31/2022]
Abstract
During the orthodontic tooth movement, cells in periodontal ligament could differentiate into osteoblasts to synthesize alveolar bone as well as affect the proliferation, migration and differentiation of mesenchymal stem cells, which also contribute to bone remodeling. However, the mechanism is still largely elusive. Here, we evaluated the expression of CREB at the tension site of mouse periodontal ligament under orthodontic mechanical strain and in the cyclic tension strain treated human periodontal ligament cells. Then, through gain and loss of function analysis, we revealed that CREB in PDLCs promotes SDF-1 and FGF2 secretion, which enhance the migration and osteoblastic differentiation of BMSCs. We further discovered that CREB transcriptionally activates FGF2 and SDF-1 expressions by binding to the promoter regions.In conclusion, this study confirms that CREB is an upregulated gene in periodontal ligament under orthodontic tension strain stimulation and plays an important role in regulating BMSCs' physiological activity in orthodontic tension strain-induced bone formation.
Collapse
Affiliation(s)
- Maolin Chang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Heng Lin
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Haidi Fu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jie Wang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Yang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ziqiu Wan
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guangli Han
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
14
|
Ni S, Xiong XB, Ni XY. MgCl2 promotes mouse mesenchymal stem cell osteogenic differentiation by activating the p38/Osx/Runx2 signaling pathway. Mol Med Rep 2020; 22:3904-3910. [PMID: 32901870 PMCID: PMC7533493 DOI: 10.3892/mmr.2020.11487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 07/15/2020] [Indexed: 12/30/2022] Open
Abstract
Magnesium, an important inorganic mineral component in bones, enhances osteoblast adhesion and osteogenic gene expression. Mg2+‑containing hydroxyapatite promotes mouse mesenchymal stem cell (MMSC) osteogenic differentiation. In the present study, MMSCs were cultured in media containing different concentrations of MgCl2 (0 and 20 mM) for different time periods. Western blotting and reverse transcription‑quantitative PCR were performed to determine the expression levels of phosphorylated (p)‑p38 mitogen‑activated protein kinase (MAPK), the osteoblast‑specific transcription factor Osterix (Osx), runt‑related transcription factor 2 (Runx2), and p38 downstream genes, such as 27 kDa heat shock protein (hsp27), activating transcription factor 4 (Atf4), myocyte enhancer factor 2C (Mef2c) and CCAAT/enhancer‑binding protein homologous protein (Ddit3). The facilitatory effect of MgCl2 on MMSC osteogenic differentiation was assessed via Alizarin Red staining. The results suggested that MgCl2 increased p38 phosphorylation compared with the control group. Downstream genes of the p38 signaling pathway, including Osx and Runx2, as well as several osteogenesis‑associated downstream target genes, including Hsp27, Atf4, Ddit3 and Mef2c, were significantly upregulated in the Mg2+‑treated group compared with the control group. The increased osteogenic differentiation in the Mg2+‑treated group was significantly attenuated in MMSCs treated with SB203580, a specific inhibitor of the p38 signaling pathway. The results suggested that appropriate concentrations of MgCl2 promoted MMSC osteogenic differentiation via regulation of the p38/Osx/Runx2 signaling pathway.
Collapse
Affiliation(s)
- Su Ni
- Medical Research Center, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Xin-Bo Xiong
- Shenzhen Key Laboratory of Special Functional Materials, College of Materials, Shenzhen University, Shenzhen, Guangdong 518086, P.R. China
| | - Xin-Ye Ni
- Medical Research Center, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
15
|
Chen WX, Liu HH, Li RX, Mammadov G, Wang JJ, Liu FF, Samadli S, Wu YF, Zhang DD, Luo HH, Hu P. C-type natriuretic peptide stimulates osteoblastic proliferation and collagen-X expression but suppresses fibroblast growth factor-23 expression in vitro. Pediatr Rheumatol Online J 2020; 18:46. [PMID: 32517762 PMCID: PMC7285564 DOI: 10.1186/s12969-020-00441-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/03/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The effects of C-type natriuretic peptide (CNP) and fibroblast growth factor (FGF)-23 appear to oppose each other during the process of bone formation, whereas few studies exist on the interaction between CNP and FGF-23. The main objective of the present study is to probe whether CNP is directly responsible for the regulation of osteoblast or via antagonizing FGF-23. METHODS Osteoblasts were cultured in the absence or presence of CNP (0, 10, and 100 pmol/L) for 24 h, 48 h and 72 h, respectively. RESULTS The findings of the present study indicated that: (1) CNP significantly stimulated osteoblastic proliferation and collagen (Col)-X expression; (2) both osteoblastic (osteocalcin, procollagen type I carboxy-terminal propeptide, total alkaline phosphatase and bone-specific alkaline phosphatase) and osteolytic (tartrate-resistant acid phosphatase and cross-linked carboxyterminal telopeptide of type I collagen) bone turnover biomarkers were up-regulated by CNP in osteoblasts; (3) FGF-23 mRNA and protein were significantly down-regulated at 24 h by CNP in osteoblasts, but the expression of FGF receptor-1/Klotho had no significant change. CONCLUSIONS CNP stimulates osteoblastic proliferation and Col-X expression via the down-regulation of FGF-23 possibly in vitro. However, the specific mechanisms of the interaction between CNP and FGF-23 in osteoblasts are still unclear according to our findings. A further study on osteoblasts cultured with CNP and FGF-23 inhibitor will be undertaken in our laboratory.
Collapse
Affiliation(s)
- Wei Xia Chen
- grid.412679.f0000 0004 1771 3402Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, Anhui Province 230032 PR China
| | - Hui Hui Liu
- grid.412679.f0000 0004 1771 3402Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, Anhui Province 230032 PR China
| | - Rui Xue Li
- grid.412679.f0000 0004 1771 3402Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, Anhui Province 230032 PR China
| | - Goshgar Mammadov
- grid.412679.f0000 0004 1771 3402Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, Anhui Province 230032 PR China
| | - Jing Jing Wang
- grid.412679.f0000 0004 1771 3402Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, Anhui Province 230032 PR China
| | - Fei Fei Liu
- grid.412679.f0000 0004 1771 3402Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, Anhui Province 230032 PR China
| | - Sama Samadli
- grid.412679.f0000 0004 1771 3402Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, Anhui Province 230032 PR China
| | - Yang Fang Wu
- grid.412679.f0000 0004 1771 3402Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, Anhui Province 230032 PR China
| | - Dong Dong Zhang
- grid.412679.f0000 0004 1771 3402Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, Anhui Province 230032 PR China
| | - Huang Huang Luo
- grid.412679.f0000 0004 1771 3402Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, Anhui Province 230032 PR China
| | - Peng Hu
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, No. 218 Ji-Xi Road, Hefei, Anhui Province, 230032, PR China.
| |
Collapse
|
16
|
MicroRNA-16, via FGF2 Regulation of the ERK/MAPK Pathway, Is Involved in the Magnesium-Promoted Osteogenic Differentiation of Mesenchymal Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3894926. [PMID: 32411326 PMCID: PMC7201663 DOI: 10.1155/2020/3894926] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/16/2020] [Indexed: 12/26/2022]
Abstract
microRNAs (miRNAs) participate in the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). However, few reports have discussed the effect of miRNAs on the magnesium chloride (MgCl2)-induced promotion of osteogenic differentiation of BMSCs, a process involved in the healing of bone tissue. As determined in the present investigation, MgCl2 decreased miR-16 levels; increased levels of fibroblast growth factor 2 (FGF2), p-p38, and p-ERK; and promoted the osteogenic differentiation of BMSCs. Enhancement of miR-16 levels by an miR-16 mimic blocked these MgCl2-induced changes. Moreover, luciferase reporter assays confirmed that miR-16 binds to the 3'UTR region of FGF2 mRNA. Down-regulation of FGF2 blocked the MgCl2-induced increases of p-p38 and p-ERK and the promotion of the osteogenic differentiation of BMSCs. Furthermore, over-expression of miR-16 attenuated the MgCl2-induced overproduction of p-p38 and p-ERK1/2 and the high levels of osteogenic differentiation, effects that were reversed by elevated expression of FGF2. In summary, the present findings provide a mechanism by which miR-16 regulates MgCl2-induced promotion of osteogenic differentiation by targeting FGF2-mediated activation of the ERK/MAPK pathway.
Collapse
|
17
|
Marx D, Rahimnejad Yazdi A, Papini M, Towler M. A review of the latest insights into the mechanism of action of strontium in bone. Bone Rep 2020; 12:100273. [PMID: 32395571 PMCID: PMC7210412 DOI: 10.1016/j.bonr.2020.100273] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/09/2020] [Accepted: 04/16/2020] [Indexed: 02/08/2023] Open
Abstract
Interest in strontium (Sr) has persisted over the last three decades due to its unique mechanism of action: it simultaneously promotes osteoblast function and inhibits osteoclast function. While this mechanism of action is strongly supported by in vitro studies and small animal trials, recent large-scale clinical trials have demonstrated that orally administered strontium ranelate (SrRan) may have no anabolic effect on bone formation in humans. Yet, there is a strong correlation between Sr accumulation in bone and reduced fracture risk in post-menopausal women, suggesting Sr acts via a purely physiochemical mechanism to enhance bone strength. Conversely, the local administration of Sr with the use of modified biomaterials has been shown to enhance bone growth, osseointegration and bone healing at the bone-implant interface, to a greater degree than Sr-free materials. This review summarizes current knowledge of the main cellular and physiochemical mechanisms that underly Sr's effect in bone, which center around Sr's similarity to calcium (Ca). We will also summarize the main controversies in Sr research which cast doubt on the 'dual-acting mechanism'. Lastly, we will explore the effects of Sr-modified bone-implant materials both in vitro and in vivo, examining whether Sr may act via an alternate mechanism when administered locally.
Collapse
Affiliation(s)
- Daniella Marx
- Department of Biomedical Engineering, Ryerson University, Toronto M5B 2K3, Ontario, Canada.,Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto M5B 1W8, Ontario, Canada
| | - Alireza Rahimnejad Yazdi
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto M5B 1W8, Ontario, Canada.,Department of Mechanical Engineering, Ryerson University, Toronto M5B 2K3, Ontario, Canada
| | - Marcello Papini
- Department of Biomedical Engineering, Ryerson University, Toronto M5B 2K3, Ontario, Canada.,Department of Mechanical Engineering, Ryerson University, Toronto M5B 2K3, Ontario, Canada
| | - Mark Towler
- Department of Biomedical Engineering, Ryerson University, Toronto M5B 2K3, Ontario, Canada.,Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto M5B 1W8, Ontario, Canada.,Department of Mechanical Engineering, Ryerson University, Toronto M5B 2K3, Ontario, Canada
| |
Collapse
|
18
|
Shafaei H, Kalarestaghi H. Adipose-derived stem cells: An appropriate selection for osteogenic differentiation. J Cell Physiol 2020; 235:8371-8386. [PMID: 32239731 DOI: 10.1002/jcp.29681] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are a major component of various forms of tissue engineering. MSCs have self-renewal and multidifferential potential. Osteogenic differentiation of MSCs is an area of attention in bone regeneration. One form of MSCs are adipose-derived stem cells (ASCs), which can be simply harvested and differentiated into several cell lineages, such as chondrocytes, adipocytes, or osteoblasts. Due to special properties, ASCs are frequently used in vitro and in vivo bone regeneration. Identifying factors involved in osteogenic differentiation of ASCs is important for better understanding the mechanism of osteogenic differentiation. Different methods are used to stimulate osteogenesis of ASCs in literature, including common osteogenic media, growth factors, hormones, hypoxia, mechanical and chemical stimuli, genetic modification, and nanotechnology. This review article provides an overview describing the isolation procedure, characterization, properties, current methods for osteogenic differentiation of ASCs, and their basic biological mechanism.
Collapse
Affiliation(s)
- Hajar Shafaei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Kalarestaghi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran.,Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences and Pathology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
19
|
Kosowska K, Domalik-Pyzik P, Krok-Borkowicz M, Chłopek J. Polylactide/Hydroxyapatite Nonwovens Incorporated into Chitosan/Graphene Materials Hydrogels to Form Novel Hierarchical Scaffolds. Int J Mol Sci 2020; 21:ijms21072330. [PMID: 32230916 PMCID: PMC7178071 DOI: 10.3390/ijms21072330] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022] Open
Abstract
In this study, hierarchical, cylindrical scaffolds based on polylactide (PLA) microfibers incorporated into chitosan (CS) hydrogel were prepared for potential use in bone tissue engineering. PLA nonwovens modified with hydroxyapatite particles (HAp) were obtained using the electrospinning method. Then, three-dimensional scaffolds were created by rolling up the nonwovens and immersing them in CS-based solutions with graphene oxide (GO) or reduced graphene oxide (rGO) dispersed in the polymer matrix. Hydrogels were cross-linked using a novel freezing-thawing-gelling method. A broad spectrum of research methods was applied in order to thoroughly characterize both the nanofillers and the composite systems: scanning electron microscopy, X-ray photoelectron spectroscopy, X-ray diffractometry, attenuated total reflection Fourier transform infrared spectroscopy, rheological and mechanical testing, as well as the assessment of chemical stability, bioactivity and cytocompatibility.
Collapse
|
20
|
Wang N, Niger C, Li N, Richards GO, Skerry TM. Cross-Species RNA-Seq Study Comparing Transcriptomes of Enriched Osteocyte Populations in the Tibia and Skull. Front Endocrinol (Lausanne) 2020; 11:581002. [PMID: 33071985 PMCID: PMC7543096 DOI: 10.3389/fendo.2020.581002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/21/2020] [Indexed: 12/17/2022] Open
Abstract
Local site-specific differences between bones in different regions of the skeleton account for their different properties and functions. To identify mechanisms behind these differences, we have performed a cross-species study comparing RNA transcriptomes of cranial and tibial osteocytes, from bones with very different primary functions and physiological responses, collected from the same individual mouse, rat, and rhesus macaque. Bioinformatic analysis was performed to identify 32 genes changed in the same direction between sites and shared across all three species. Several well-established key genes in bone growth and remodeling were upregulated in the tibias of all three species (BMP7, DKK1, FGF1, FRZB, SOST). Many of them associate or crosstalk with the Wnt signaling pathway. These results suggest Wnt signaling-related candidates for different control of regulatory mechanisms in bone homeostasis in the skull and tibia and indicate a different balance between genetically determined structure and feedback mechanisms to strains induced by mechanical loading at the different sites.
Collapse
Affiliation(s)
- Ning Wang
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, United Kingdom
| | - Corinne Niger
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, United Kingdom
| | - Nan Li
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - Gareth O. Richards
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, United Kingdom
| | - Tim M. Skerry
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, United Kingdom
- *Correspondence: Tim M. Skerry
| |
Collapse
|
21
|
Yuan X, Liu M, Cao X, Yang S. Ciliary IFT80 regulates dental pulp stem cells differentiation by FGF/FGFR1 and Hh/BMP2 signaling. Int J Biol Sci 2019; 15:2087-2099. [PMID: 31592124 PMCID: PMC6775288 DOI: 10.7150/ijbs.27231] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 02/19/2019] [Indexed: 01/09/2023] Open
Abstract
Primary cilia and intraflagellar transport (IFT) proteins control a wide variety of processes during development and tissue homeostasis. However, their potential roles in the regulation of stem cell differentiation and tooth development remain elusive. Here, we uncovered the critical roles of ciliary IFT80 in cilia formation and differentiation of dental pulp stem cells (DPSCs). IFT80-deficient DPSCs showed reduced fibroblast growth factor receptor 1 (FGFR1) expression, leading to the disruption of FGF2-FGFR1 signaling. We found, during DPSC differentiation, FGF2-FGFR1 signaling induces stress fiber rearrangement to promote cilia elongation, meanwhile stimulates PI3K-AKT signaling to aid Hh/bone morphogenetic protein 2 (BMP2) signaling activation. These signaling pathways and their coupling were disrupted in IFT80-deficient DPSCs, causing impaired differentiation. Our findings revealed a novel mechanism that ciliary protein regulates the odontogenic differentiation of DPSCs through FGF/FGFR1 and Hh/BMP2 signaling.
Collapse
Affiliation(s)
- Xue Yuan
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY, United States
| | - Min Liu
- Department of Anatomy & Cell Biology, School of Dental Medicine, University of Pennsylvania, PA, United States
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shuying Yang
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY, United States
- Department of Anatomy & Cell Biology, School of Dental Medicine, University of Pennsylvania, PA, United States
| |
Collapse
|
22
|
Alvarez C, Monasterio G, Cavalla F, Córdova LA, Hernández M, Heymann D, Garlet GP, Sorsa T, Pärnänen P, Lee HM, Golub LM, Vernal R, Kantarci A. Osteoimmunology of Oral and Maxillofacial Diseases: Translational Applications Based on Biological Mechanisms. Front Immunol 2019; 10:1664. [PMID: 31379856 PMCID: PMC6657671 DOI: 10.3389/fimmu.2019.01664] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 07/03/2019] [Indexed: 12/23/2022] Open
Abstract
The maxillofacial skeleton is highly dynamic and requires a constant equilibrium between the bone resorption and bone formation. The field of osteoimmunology explores the interactions between bone metabolism and the immune response, providing a context to study the complex cellular and molecular networks involved in oro-maxillofacial osteolytic diseases. In this review, we present a framework for understanding the potential mechanisms underlying the immuno-pathobiology in etiologically-diverse diseases that affect the oral and maxillofacial region and share bone destruction as their common clinical outcome. These otherwise different pathologies share similar inflammatory pathways mediated by central cellular players, such as macrophages, T and B cells, that promote the differentiation and activation of osteoclasts, ineffective or insufficient bone apposition by osteoblasts, and the continuous production of osteoclastogenic signals by immune and local stromal cells. We also present the potential translational applications of this knowledge based on the biological mechanisms involved in the inflammation-induced bone destruction. Such applications can be the development of immune-based therapies that promote bone healing/regeneration, the identification of host-derived inflammatory/collagenolytic biomarkers as diagnostics tools, the assessment of links between oral and systemic diseases; and the characterization of genetic polymorphisms in immune or bone-related genes that will help diagnosis of susceptible individuals.
Collapse
Affiliation(s)
- Carla Alvarez
- Forsyth Institute, Cambridge, MA, United States
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Gustavo Monasterio
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Franco Cavalla
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Luis A. Córdova
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, San Jose's Hospital and Clínica Las Condes, Universidad de Chile, Santiago, Chile
| | - Marcela Hernández
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Dominique Heymann
- INSERM, UMR 1232, LabCT, CRCINA, Institut de Cancérologie de l'Ouest, Université de Nantes, Université d'Angers, Saint-Herblain, France
| | - Gustavo P. Garlet
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Timo Sorsa
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
- Department of Oral Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Pirjo Pärnänen
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Hsi-Ming Lee
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Lorne M. Golub
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Dentistry Unit, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | | |
Collapse
|
23
|
Dianat-Moghadam H, Teimoori-Toolabi L. Implications of Fibroblast Growth Factors (FGFs) in Cancer: From Prognostic to Therapeutic Applications. Curr Drug Targets 2019; 20:852-870. [DOI: 10.2174/1389450120666190112145409] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/01/2019] [Accepted: 01/02/2019] [Indexed: 12/22/2022]
Abstract
Fibroblast growth factors (FGFs) are pleiotropic molecules exerting autocrine, intracrine
and paracrine functions via activating four tyrosine kinase FGF receptors (FGFR), which further trigger
a variety of cellular processes including angiogenesis, evasion from apoptosis, bone formation,
embryogenesis, wound repair and homeostasis. Four major mechanisms including angiogenesis, inflammation,
cell proliferation, and metastasis are active in FGF/FGFR-driven tumors. Furthermore,
gain-of-function or loss-of-function in FGFRs1-4 which is due to amplification, fusions, mutations,
and changes in tumor–stromal cells interactions, is associated with the development and progression
of cancer. Although, the developed small molecule or antibodies targeting FGFR signaling offer immense
potential for cancer therapy, emergence of drug resistance, activation of compensatory pathways
and systemic toxicity of modulators are bottlenecks in clinical application of anti-FGFRs. In this
review, we present FGF/FGFR structure and the mechanisms of its function, as well as cross-talks
with other nodes and/or signaling pathways. We describe deregulation of FGF/FGFR-related mechanisms
in human disease and tumor progression leading to the presentation of emerging therapeutic approaches,
resistance to FGFR targeting, and clinical potentials of individual FGF family in several
human cancers. Additionally, the underlying biological mechanisms of FGF/FGFR signaling, besides
several attempts to develop predictive biomarkers and combination therapies for different cancers
have been explored.
Collapse
Affiliation(s)
- Hassan Dianat-Moghadam
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Ladan Teimoori-Toolabi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
24
|
Wang X, Liu Q, Chen W, Liu L. FGF adsorbed mesoporous bioactive glass with larger pores in enhancing bone tissue engineering. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2019; 30:48. [PMID: 30982116 DOI: 10.1007/s10856-019-6252-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 04/04/2019] [Indexed: 06/09/2023]
Abstract
Mesoporous bioactive glass (MBG) is performed as a bone tissue engineering material because of its good bioactivity, biocompatibility and osteoinducion characteristics. Here, we propose MBG with larger pores (MBG-L) adsorbed fibroblast growth factor (FGF) to facilitate osteoblast differentiation and matrix mineralization. Specifically, we observed that MBG-L promotes calcium deposit precipitation in vitro. In addition, adhesion, proliferation, differentiation and matrix mineralization were promoted after osteoblast cultured on MBG-L/FGF. Interestingly, we found that the transcriptional activity of the critical transcription factor Runx2 was increased through MAPK pathway after osteoblast cultured on MBG-L/FGF. Support for this result, we found that the expression of osteoblastic marker genes, Osteocalcin (Ocn), Osteopontin (Opn), and Runx2 were increased. Thus, our findings provided that MBG-L/FGF could be a promising new material in bone tissue engineering.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, 410073, Changsha, Hunan, P. R. China.
| | - Qianqian Liu
- Department of Biochemistry, School of Life Sciences, Central South University, 410013, Changsha, Hunan, P. R. China
| | - Wei Chen
- Department of Life Sciences, College of Life Sciences, Hunan Normal University, 410081, Changsha, Hunan, P. R. China
| | - Long Liu
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, 410073, Changsha, Hunan, P. R. China
| |
Collapse
|
25
|
MORILLO CMR, SLONIAK MC, GONÇALVES F, VILLAR CC. Efficacy of stem cells on bone consolidation of distraction osteogenesis in animal models: a systematic review. Braz Oral Res 2018; 32:e83. [DOI: 10.1590/1807-3107bor-2018.vol32.0083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/27/2018] [Indexed: 12/22/2022] Open
|
26
|
Liu XY, Li X, Bai MR, Chen X, Wang CL, Xie J, Ye L. FGF-7 Dictates Osteocyte Cell Processes Through Beta-Catenin Transduction. Sci Rep 2018; 8:14792. [PMID: 30287900 PMCID: PMC6172271 DOI: 10.1038/s41598-018-33247-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/26/2018] [Indexed: 02/05/2023] Open
Abstract
It is well recognized that osteocytes communicate with each other via gap junctions and that connxin43 (Cx43) shows its great potential in gap junction for the contribution enabling transmission of small molecules and operating in an autocrine/a paracrine manner. Fibroblast growth factors (FGFs) play significant roles in new bone formation and adult bone remodeling, and FGF signaling is regulated by the precise spatiotemporal approaches. However, the influence of FGF7 on osteocyte cell processes is not well elucidated. In this study, we aimed to examine the impact of FGF7 on osteocyte cell processes by characterizing the expression of Cx43 and to reveal the underlying mechanism regulating this cell process. We first found that the mRNA level of FGF7 was higher relative to other FGF family members both in osteocytes cell line (MLO-Y4) and bone tissue. We then demonstrated that FGF7 could increase the expression of Cx43 in osteocytes and promote the cell processes in the form of gap junctions between osteocytes. This modulation was due to the FGF7-induced cytoplasmic accumulation and resultant nuclear translocation of β-catenin. Our results could help us to further understand the importance of FGF7 on bone cell behavior and bone physiology and even pathology.
Collapse
Affiliation(s)
- Xiao-Yu Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ming-Ru Bai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xia Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Cheng-Lin Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Ling Ye
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
27
|
Robu AC, Popescu L, Seidler DG, Zamfir AD. Chip-based high resolution tandem mass spectrometric determination of fibroblast growth factor-chondroitin sulfate disaccharides noncovalent interaction. JOURNAL OF MASS SPECTROMETRY : JMS 2018; 53:624-634. [PMID: 29676520 DOI: 10.1002/jms.4193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/04/2018] [Accepted: 04/11/2018] [Indexed: 06/08/2023]
Abstract
Fibroblast growth factor-2 (FGF-2) is involved in wound healing and embryonic development. Glycosaminoglycans (GAGs), the major components of the extracellular matrix (ECM), play fundamental roles at this level. FGF-GAG noncovalent interactions are in the focus of research, due to their influence upon cell proliferation and tissue regeneration. Lately, high resolution mass spectrometry (MS) coupled with chip-nanoelectrospray (nanoESI) contributed a significant progress in glycosaminoglycomics by discoveries related to novel species and their characterization. We have employed a fully automated chip-nanoESI coupled to a quadrupole time-of-flight (QTOF) MS for assessing FGF-GAG noncovalent complexes. For the first time, a CS disaccharide was involved in a binding assay with FGF-2. The experiments were conducted in 10 mM ammonium acetate/formic acid, pH 6.8, by incubating FGF-2 and CS in buffer. The detected complexes were characterized by top-down in tandem MS (MS/MS) using collision induced-dissociation (CID). CID MS/MS provided data showing for the first time that the binding process occurs via the sulfate group located at C4 in GalNAc. This study has demonstrated that chip-MS may generate reliable data upon the formation of GAG-protein complexes and their structure. Biologically, the findings are relevant for studies focused on the identification of the active domains in longer GAG chains.
Collapse
Affiliation(s)
- Adrian C Robu
- Mass Spectrometry Laboratory, National Institute for Research and Development in Electrochemistry and Condensed Matter, Plautius Andronescu Str. 1, RO-300224, Timisoara, Romania
- Faculty of Physics, West University of Timisoara, Blvd. Vasile Parvan 4, RO-300223, Timisoara, Romania
| | - Laurentiu Popescu
- Mass Spectrometry Laboratory, National Institute for Research and Development in Electrochemistry and Condensed Matter, Plautius Andronescu Str. 1, RO-300224, Timisoara, Romania
- Faculty of Physics, West University of Timisoara, Blvd. Vasile Parvan 4, RO-300223, Timisoara, Romania
| | - Daniela G Seidler
- Department of Gastroentero-, Hepato-, and Endocrinology I3, Hannover Medical School, EB2/R3110, Carl-Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Alina D Zamfir
- Mass Spectrometry Laboratory, National Institute for Research and Development in Electrochemistry and Condensed Matter, Plautius Andronescu Str. 1, RO-300224, Timisoara, Romania
- Department of Chemical and Biological Sciences, "Aurel Vlaicu" University of Arad, Revolutiei Blvd. 77, RO-310130, Arad, Romania
| |
Collapse
|
28
|
Matrix metalloproteinase-13: A special focus on its regulation by signaling cascades and microRNAs in bone. Int J Biol Macromol 2018; 109:338-349. [DOI: 10.1016/j.ijbiomac.2017.12.091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 12/15/2017] [Accepted: 12/17/2017] [Indexed: 01/03/2023]
|
29
|
Impairment of PTX3 expression in osteoblasts: a key element for osteoporosis. Cell Death Dis 2017; 8:e3125. [PMID: 29022895 PMCID: PMC5682679 DOI: 10.1038/cddis.2017.514] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/19/2017] [Accepted: 08/30/2017] [Indexed: 01/21/2023]
Abstract
Pentraxin 3 (PTX3) is a multifunctional glycoprotein regulating inflammatory response, cell proliferation and migration and deposition and remodelling of the extracellular matrix by a variety of cells. In this study, we investigated the possible role of PTX3 in bone homeostasis. To this end, we compared the expression and function of PTX3 in human osteoblasts of osteoporotic, osteoarthritic patients and young subjects not affected by bone diseases. Immunohistochemical analysis performed on bone head biopsies showed a close association between bone health and the number of osteoblasts expressing PTX3. Noteworthy, the proportion of PTX3-positive osteoblasts resulted to be significantly lower in osteoporotic patients compared with both young patients and osteoarthritic patients of the same age. Ex vivo culture of osteoblasts isolated from the three groups of patients confirmed in vivo observation. Specifically, we observed rare runt-related transcription factor 2 (RUNX2) immunopositive osteoblasts expressing PTX3 in cell cultures derived from osteoporotic patients and western blotting analysis showed 80% reduction of PTX3 in the corresponding culture extracts compared with young and osteoarthritic patients. The treatment of human osteoblast primary cultures derived from young patients with anti-PTX3 antibody dramatically affected osteoblast behaviour. Indeed, they lost the morphological and molecular features typical of mature osteoblasts, acquiring fibroblast-like shape and highly decreasing nuclear factor kappa-B ligand (RANKL) and RUNX2 expression. Also, the inhibition of PTX3 negatively affected osteoblast proliferation and their ability to form cell clusters and microhydroxyapatite crystals. Altogether, these results suggest a central role of PTX3 in bone homeostasis showing its involvement in osteoblast proliferation, differentiation and function.
Collapse
|
30
|
Coulson J, Bagley L, Barnouin Y, Bradburn S, Butler-Browne G, Gapeyeva H, Hogrel JY, Maden-Wilkinson T, Maier AB, Meskers C, Murgatroyd C, Narici M, Pääsuke M, Sassano L, Sipilä S, Al-Shanti N, Stenroth L, Jones DA, McPhee JS. Circulating levels of dickkopf-1, osteoprotegerin and sclerostin are higher in old compared with young men and women and positively associated with whole-body bone mineral density in older adults. Osteoporos Int 2017; 28:2683-2689. [PMID: 28585053 DOI: 10.1007/s00198-017-4104-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/23/2017] [Indexed: 12/31/2022]
Abstract
UNLABELLED Bone mineral density declines with increasing older age. We examined the levels of circulating factors known to regulate bone metabolism in healthy young and older adults. The circulating levels of dickkopf-1, osteocalcin, osteoprotegerin and sclerostin were positively associated with whole-body bone mineral density (WBMD) in older adults, despite the average WBMD being lower and circulating dickkopf-1, osteoprotegerin and sclerostin being higher in old than young. INTRODUCTION This study aims to investigate the relationship between whole-body bone mineral density (WBMD) and levels of circulating factors with known roles in bone remodelling during 'healthy' ageing. METHODS WBMD and fasting plasma concentrations of dickkopf-1, fibroblast growth factor-23, osteocalcin, osteoprotegerin, osteopontin and sclerostin were measured in 272 older subjects (69 to 81 years; 52% female) and 171 younger subjects (18-30 years; 53% female). RESULTS WBMD was lower in old than young. Circulating osteocalcin was lower in old compared with young, while dickkopf-1, osteoprotegerin and sclerostin were higher in old compared with young. These circulating factors were each positively associated with WBMD in the older adults and the relationships remained after adjustment for covariates (r values ranging from 0.174 to 0.254, all p < 0.01). In multivariate regression, the body mass index, circulating sclerostin and whole-body lean mass together accounted for 13.8% of the variation with WBMD in the older adults. In young adults, dickkopf-1 and body mass index together accounted for 7.7% of variation in WBMD. CONCLUSION Circulating levels of dickkopf-1, osteocalcin, osteoprotegerin and sclerostin are positively associated with WBMD in community-dwelling older adults, despite the average WBMD being lower and circulating dickkopf-1, osteoprotegerin and sclerostin being higher in old than young.
Collapse
Affiliation(s)
- J Coulson
- School of Healthcare Science, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester, M15GD, UK
| | - L Bagley
- School of Healthcare Science, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester, M15GD, UK
| | - Y Barnouin
- School of Healthcare Science, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester, M15GD, UK
- Baylor College of Medicine, Houston, TX, USA
| | - S Bradburn
- School of Healthcare Science, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester, M15GD, UK
| | | | - H Gapeyeva
- Institute of Sport Sciences and Physiotherapy, University of Tartu, Tartu, Estonia
| | - J-Y Hogrel
- Institut de Myologie, GH Pitié-Salpêtrière, Paris, France
| | - T Maden-Wilkinson
- School of Physical Activity and Health, Sheffield Hallam University, Sheffield, UK
| | - A B Maier
- Department of Human Movement Sciences, MOVE Research Institute, Vrij University, Amsterdam, The Netherlands
- Department of Medicine and Aged Care, The Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
| | - C Meskers
- Rehabilitation Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - C Murgatroyd
- School of Healthcare Science, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester, M15GD, UK
| | - M Narici
- Graduate Entry Medicine and Health, University of Nottingham, Nottingham, UK
| | - M Pääsuke
- Institute of Sport Sciences and Physiotherapy, University of Tartu, Tartu, Estonia
| | - L Sassano
- Unilever Discover, Colworth Park, Sharnbrook, Bedford, UK
| | - S Sipilä
- Gerontology Research Center, Department of Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - N Al-Shanti
- School of Healthcare Science, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester, M15GD, UK
| | - L Stenroth
- Gerontology Research Center, Department of Health Sciences, University of Jyväskylä, Jyväskylä, Finland
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - D A Jones
- School of Healthcare Science, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester, M15GD, UK
| | - J S McPhee
- School of Healthcare Science, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester, M15GD, UK.
| |
Collapse
|
31
|
Kuljanin M, Brown CFC, Raleigh MJ, Lajoie GA, Flynn LE. Collagenase treatment enhances proteomic coverage of low-abundance proteins in decellularized matrix bioscaffolds. Biomaterials 2017; 144:130-143. [PMID: 28829951 DOI: 10.1016/j.biomaterials.2017.08.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/03/2017] [Accepted: 08/12/2017] [Indexed: 12/12/2022]
Abstract
There is great interest in the application of advanced proteomic techniques to characterize decellularized tissues in order to develop a deeper understanding of the effects of the complex extracellular matrix (ECM) composition on the cellular response to these pro-regenerative bioscaffolds. However, the identification of proteins in ECM-derived bioscaffolds is hindered by the high abundance of collagen in the samples, which can interfere with the detection of lower-abundance constituents that may be important regulators of cell function. To address this limitation, we developed a novel multi-enzyme digestion approach using treatment with a highly-purified collagenase derived from Clostridium Histolyticum to selectively deplete collagen from ECM-derived protein extracts, reducing its relative abundance from up to 90% to below 10%. Moreover, we applied this new method to characterize the proteome of human decellularized adipose tissue (DAT), human decellularized cancellous bone (DCB), and commercially-available bovine tendon collagen (BTC). We successfully demonstrated with all three sources that collagenase treatment increased the depth of detection and enabled the identification of a variety of signaling proteins that were masked by collagen in standard digestion protocols with trypsin/LysC, increasing the number of proteins identified in the DAT by ∼2.2 fold, the DCB by ∼1.3 fold, and the BTC by ∼1.6 fold. In addition, quantitative proteomics using label-free quantification demonstrated that the DAT and DCB extracts were compositionally distinct, and identified a number of adipogenic and osteogenic proteins that were consistently more highly expressed in the DAT and DCB respectively. Overall, we have developed a new processing method that may be applied in advanced mass spectrometry studies to improve the high-throughput proteomic characterization of bioscaffolds derived from mammalian tissues. Further, our study provides new insight into the complex ECM composition of two human decellularized tissues of interest as cell-instructive platforms for regenerative medicine.
Collapse
Affiliation(s)
- Miljan Kuljanin
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Cody F C Brown
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Matthew J Raleigh
- Undergraduate Medical Education, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Gilles A Lajoie
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada.
| | - Lauren E Flynn
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada; Department of Chemical and Biochemical Engineering, Thompson Engineering Building, The University of Western Ontario, London, Ontario, N6A 5B9, Canada.
| |
Collapse
|
32
|
Zhai F, Song N, Ma J, Gong W, Tian H, Li X, Jiang C, Wang H. FGF18 inhibits MC3T3-E1 cell osteogenic differentiation via the ERK signaling pathway. Mol Med Rep 2017; 16:4127-4132. [DOI: 10.3892/mmr.2017.7088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/12/2017] [Indexed: 11/05/2022] Open
|
33
|
Wang L, Roth T, Abbott M, Ho L, Wattanachanya L, Nissenson RA. Osteoblast-derived FGF9 regulates skeletal homeostasis. Bone 2017; 98:18-25. [PMID: 28189801 PMCID: PMC8474898 DOI: 10.1016/j.bone.2016.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 12/01/2016] [Accepted: 12/10/2016] [Indexed: 11/21/2022]
Abstract
FGF9 has complex and important roles in skeletal development and repair. We have previously observed that Fgf9 expression in osteoblasts (OBs) is regulated by G protein signaling and therefore the present study was done to determine whether OB-derived FGF9 was important in skeletal homeostasis. To directly test this idea, we deleted functional expression of Fgf9 gene in OBs using a 2.3kb collagen type I promoter-driven Cre transgenic mouse line (Fgf9OB-/-). Both Fgf9 knockout (Fgf9OB-/-) and the Fgf9 floxed littermates (Fgf9fl/fl) mice were fully backcrossed and maintained in an FBV/N background. Three month old Fgf9OB-/- mice displayed a significant decrease in cancellous bone and bone formation in the distal femur and a significant decrease in cortical thickness at the TFJ. Strikingly, female Fgf9OB-/- mice did not display altered bone mass. Continuous treatment of mouse BMSCs with exogenous FGF9 inhibited mouse BMSC mineralization while acute treatment increased the proliferation of progenitors, an effect requiring the activation of Akt1. Our results suggest that mature OBs are an important source of FGF9, positively regulating skeletal homeostasis in male mice. Osteoblast-derived FGF9 may serve a paracrine role to maintain the osteogenic progenitor cell population through activation of Akt signaling.
Collapse
Affiliation(s)
- Liping Wang
- Endocrine Unit, VA Medical Center, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA
| | - Theresa Roth
- Endocrine Unit, VA Medical Center, San Francisco, CA, USA
| | - Marcia Abbott
- Endocrine Unit, VA Medical Center, San Francisco, CA, USA
| | - Linh Ho
- Endocrine Unit, VA Medical Center, San Francisco, CA, USA
| | - Lalita Wattanachanya
- Endocrine Unit, VA Medical Center, San Francisco, CA, USA; Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Thai Red Cross Society, Bangkok, Thailand; King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Robert A Nissenson
- Endocrine Unit, VA Medical Center, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
34
|
Creo AL, Thacher TD, Pettifor JM, Strand MA, Fischer PR. Nutritional rickets around the world: an update. Paediatr Int Child Health 2017; 37:84-98. [PMID: 27922335 DOI: 10.1080/20469047.2016.1248170] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Worldwide, nutritional rickets continues to be an evolving problem with several causes. This paper provides an updated literature review characterising the prevalence, aetiology, pathophysiology and treatment of nutritional rickets worldwide. A systematic review of articles on nutritional rickets from various geographical regions was undertaken. For each region, key information was extracted, including prevalence, cause of rickets specific to the region, methods of confirming the diagnosis and current treatment and preventive measures. Calcium deficiency continues to be a major cause of rickets in Africa and Asia. Vitamin D deficiency rickets is perhaps increasing in the Americas, Europe and parts of the Middle East. There continues to be a distinct presentation of calcium-predominant versus vitamin D predominant rickets, although there are overlapping features. More careful diagnosis of rickets and reporting of 25-OHD concentrations has improved accurate knowledge of rickets prevalence and better delineated the cause. Nutritional rickets continues to be an evolving and multi-factorial problem worldwide. It is on a spectrum, ranging from isolated vitamin D deficiency to isolated calcium deficiency. Specific areas which require emphasis include a consistent community approach to screening and diagnosis, vitamin D supplementation of infants and at-risk children, prevention of maternal vitamin D deficiency and the provision of calcium in areas with low calcium diets.
Collapse
Affiliation(s)
- Ana L Creo
- a Department of Pediatric and Adolescent Medicine , Mayo Clinic , Rochester , MN , USA
| | - Tom D Thacher
- b Department of Family Medicine , Mayo Clinic , Rochester , MN , USA
| | - John M Pettifor
- c Wits/SAMRC Developmental Pathways for Health Research Unit, Department of Paediatrics , University of the Witwatersrand , Johannesburg , South Africa
| | - Mark A Strand
- d Pharmacy Practice, Department of Public Health , North Dakota State University , Fargo , ND , USA
| | - Philip R Fischer
- a Department of Pediatric and Adolescent Medicine , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
35
|
Shahi M, Peymani A, Sahmani M. Regulation of Bone Metabolism. Rep Biochem Mol Biol 2017; 5:73-82. [PMID: 28367467 PMCID: PMC5346273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/14/2016] [Indexed: 06/07/2023]
Abstract
Bone is formed through the processes of endochondral and intramembranous ossification. In endochondral ossification primary mesenchymal cells differentiate to chondrocytes and then are progressively substituted by bone, while in intramembranous ossification mesenchymal stem cells (MSCs) differentiate directly into osteoblasts to form bone. The steps of osteogenic proliferation, differentiation, and bone homeostasis are controlled by various markers and signaling pathways. Bone needs to be remodeled to maintain integrity with osteoblasts, which are bone-forming cells, and osteoclasts, which are bone-degrading cells.In this review we considered the major factors and signaling pathways in bone formation; these include fibroblast growth factors (FGFs), bone morphogenetic proteins (BMPs), wingless-type (Wnt) genes, runt-related transcription factor 2 (RUNX2) and osteoblast-specific transcription factor (osterix or OSX).
Collapse
Affiliation(s)
- Maryam Shahi
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Amir Peymani
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Mehdi Sahmani
- Department of Clinical Biochemistry and Medical Genetics, Cellular and Molecular Research Center, Qazvin University of
Medical Sciences, Qazvin, Iran.
| |
Collapse
|
36
|
Xia J, Sheng W, Pei L, Li N, Zhang Z, Wang J, Zu J, Wang N, Wang D. Effects of unfractionated heparin and rivaroxaban on the expression of heparanase and fibroblast growth factor 2 in human osteoblasts. Mol Med Rep 2017; 16:361-366. [DOI: 10.3892/mmr.2017.6570] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 03/13/2017] [Indexed: 11/06/2022] Open
|
37
|
Simann M, Le Blanc S, Schneider V, Zehe V, Lüdemann M, Schütze N, Jakob F, Schilling T. Canonical FGFs Prevent Osteogenic Lineage Commitment and Differentiation of Human Bone Marrow Stromal Cells Via ERK1/2 Signaling. J Cell Biochem 2016; 118:263-275. [PMID: 27305863 DOI: 10.1002/jcb.25631] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/14/2016] [Indexed: 12/21/2022]
Abstract
Controlling the adipo-osteogenic lineage decision of trabecular human bone marrow stromal cells (hBMSCs) in favor of osteogenesis represents a promising approach for osteoporosis therapy and prevention. Previously, Fibroblast Growth Factor 1 (FGF1) and its subfamily member FGF2 were scored as leading candidates to exercise control over skeletal precursor commitment and lineage decision albeit literature results are highly inconsistent. We show here that FGF1 and 2 strongly prevent the osteogenic commitment and differentiation of hBMSCs. Mineralization of extracellular matrix (ECM) and mRNA expression of osteogenic marker genes Alkaline Phosphatase (ALP), Collagen 1A1 (COL1A1), and Integrin-Binding Sialoprotein (IBSP) were significantly reduced. Furthermore, master regulators of osteogenic commitment like Runt-Related Transcription Factor 2 (RUNX2) and Bone Morphogenetic Protein 4 (BMP4) were downregulated. When administered under adipogenic culture conditions, canonical FGFs did not support osteogenic marker expression. Moreover despite the presence of osteogenic differentiation factors, FGFs even disabled the pro-osteogenic lineage decision of pre-differentiated adipocytic cells. In contrast to FGF Receptor 2 (FGFR2), FGFR1 was stably expressed throughout osteogenic and adipogenic differentiation and FGF addition. Moreover, FGFR1 and Extracellular Signal-Regulated Kinases 1 and 2 (ERK1/2) were found to be responsible for underlying signal transduction using respective inhibitors. Taken together, we present new findings indicating that canonical FGFR-ERK1/2 signaling entrapped hBMSCs in a pre-committed state and arrested further maturation of committed precursors. Our results might aid in unraveling and controlling check points relevant for ageing-associated aberrant adipogenesis with consequences for the treatment of degenerative diseases such as osteoporosis and for skeletal tissue engineering strategies. J. Cell. Biochem. 118: 263-275, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Meike Simann
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Solange Le Blanc
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Verena Schneider
- Chair Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Viola Zehe
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Martin Lüdemann
- Orthopedic Department König-Ludwig-Haus, Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Norbert Schütze
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Franz Jakob
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| | - Tatjana Schilling
- Department of Orthopedics, Orthopedic Center for Musculoskeletal Research, University of Würzburg, Würzburg, Germany
| |
Collapse
|
38
|
Zou L, Chen Q, Quanbeck Z, Bechtold JE, Kaufman DS. Angiogenic activity mediates bone repair from human pluripotent stem cell-derived osteogenic cells. Sci Rep 2016; 6:22868. [PMID: 26980556 PMCID: PMC4793227 DOI: 10.1038/srep22868] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/22/2016] [Indexed: 02/06/2023] Open
Abstract
Human pluripotent stem cells provide a standardized resource for bone repair. However, criteria to determine which exogenous cells best heal orthopedic injuries remain poorly defined. We evaluated osteogenic progenitor cells derived from both human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs). Phenotypic and genotypic analyses demonstrated that these hESCs/hiPSCs are similar in their osteogenic differentiation efficiency and they generate osteogenic cells comparable to osteogenic cells derived from mesenchymal stromal cells (BM-MSCs). However, expression of angiogenic factors, such as vascular endothelial growth factor and basic fibroblast growth factor in these osteogenic progenitor cells are markedly different, suggesting distinct pro-angiogenic potential of these stem cell derivatives. Studies to repair a femur non-union fracture demonstrate only osteogenic progenitor cells with higher pro-angiogenic potential significantly enhance bone repair in vivo. Together, these studies highlight a key role of pro-angiogenic potential of transplanted osteogenic cells for effective cell-mediated bone repair.
Collapse
Affiliation(s)
- Li Zou
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Qingshan Chen
- Excelen Center for Bone &Joint Research and Education, Minneapolis, MN, 55415, USA
| | - Zachary Quanbeck
- Excelen Center for Bone &Joint Research and Education, Minneapolis, MN, 55415, USA
| | - Joan E Bechtold
- Excelen Center for Bone &Joint Research and Education, Minneapolis, MN, 55415, USA
| | - Dan S Kaufman
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
39
|
Mattinzoli D, Rastaldi MP, Ikehata M, Armelloni S, Pignatari C, Giardino LA, Li M, Alfieri CM, Regalia A, Riccardi D, Messa P. FGF23-regulated production of Fetuin-A (AHSG) in osteocytes. Bone 2016; 83:35-47. [PMID: 26476373 DOI: 10.1016/j.bone.2015.10.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/24/2015] [Accepted: 10/13/2015] [Indexed: 12/17/2022]
Abstract
INTRODUCTION AHSG, a serum glycoprotein with recognized anti-calcification activity, has also been suggested to modulate both bone formation and resorption. Though the bulk of AHSG is mostly synthesized in the liver, it has been claimed that also bone cells might produce it. However, the extent of the bone AHSG production and the potential controlling factors remain to be definitively proven. A relevant number of studies support the notion that FGF23, a bone-derived hormone, not only regulates the most important mineral metabolism (MM) related factors (phosphate, parathyroid hormone, vitamin D, etc.), but might be also involved in cardiovascular (CV) outcome, both in chronic kidney disease (CKD) patients and in the general population. Furthermore, in addition to some direct autocrine and paracrine effects in bone, FGF23 has been suggested to interact with AHSG. In this study we investigated if AHSG is really produced by bone cells, and if its bone production is related and/or controlled by FGF23, using cultured bone cells, according to a new method recently published by our group. RESULTS Our data show that AHSG is consistently produced in osteocytes and to a far lesser extent in osteoblasts. Both FGF23 addition to the culture medium and its over-expression in osteocytes were associated with a consistent increase of both AHSG mRNA and protein, while FGF23 silencing was followed by opposite effects. Though most of these results were largely affected by the blockage of FGF23 receptors, the role of these receptors in the different experimental sets is still not completely clarified. In addition, we found that FGF23 and AHSG proteins co-localized both in cytoplasm and nucleus, which suggests a possible reciprocal interactivity. CONCLUSIONS Our data not only confirm that AHSG is produced in bone, mainly in osteocytes, but show for the first time that its production is modulated by FGF23. Since both proteins play important roles in the bone and cardiovascular pathology, these results add new pieces to the puzzling relationship between bone and vascular pathology, in particular in CKD patients, prompting future investigations in this field.
Collapse
Affiliation(s)
- D Mattinzoli
- Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, via Pace 9, 20122 Milano, Italy.
| | - M P Rastaldi
- Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, via Pace 9, 20122 Milano, Italy.
| | - M Ikehata
- Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, via Pace 9, 20122 Milano, Italy.
| | - S Armelloni
- Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, via Pace 9, 20122 Milano, Italy.
| | - C Pignatari
- Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, via Pace 9, 20122 Milano, Italy.
| | - L A Giardino
- Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, via Pace 9, 20122 Milano, Italy.
| | - M Li
- Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, via Pace 9, 20122 Milano, Italy.
| | - C M Alfieri
- Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, via Pace 9, 20122 Milano, Italy.
| | - A Regalia
- Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, via Pace 9, 20122 Milano, Italy.
| | - D Riccardi
- Division of Pathophysiology and Repair, School of Biosciences, Cardiff University, Cardiff, UK.
| | - P Messa
- Renal Research Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico & Fondazione D'Amico per la Ricerca sulle Malattie Renali, via Pace 9, 20122 Milano, Italy.
| |
Collapse
|
40
|
Fernández-Arroyo S, Huete-Toral F, Pérez de Lara MJ, de la Luz Cádiz-Gurrea M, Legeai-Mallet L, Micol V, Segura-Carretero A, Joven J, Pintor J. The impact of polyphenols on chondrocyte growth and survival: a preliminary report. Food Nutr Res 2015; 59:29311. [PMID: 26445212 PMCID: PMC4595466 DOI: 10.3402/fnr.v59.29311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/08/2015] [Accepted: 09/08/2015] [Indexed: 12/11/2022] Open
Abstract
Background Imbalances in the functional binding of fibroblast growth factors (FGFs) to their receptors (FGFRs) have consequences for cell proliferation and differentiation that in chondrocytes may lead to degraded cartilage. The toxic, proinflammatory, and oxidative response of cytokines and FGFs can be mitigated by dietary polyphenols. Objective We explored the possible effects of polyphenols in the management of osteoarticular diseases using a model based on the transduction of a mutated human FGFR3 (G380R) in murine chondrocytes. This mutation is present in most cases of skeletal dysplasia and is responsible for the overexpression of FGFR3 that, in the presence of its ligand, FGF9, results in toxic effects leading to altered cellular growth. Design Different combinations of dietary polyphenols derived from plant extracts were assayed in FGFR3 (G380R) mutated murine chondrocytes, exploring cell survival, chloride efflux, extracellular matrix (ECM) generation, and grade of activation of mitogen-activated protein kinases. Results Bioactive compounds from Hibiscus sabdariffa reversed the toxic effects of FGF9 and restored normal growth, suggesting a probable translation to clinical requests in humans. Indeed, these compounds activated the intracellular chloride efflux, increased ECM generation, and stimulated cell proliferation. The inhibition of mitogen-activated protein kinase phosphorylation was interpreted as the main mechanism governing these beneficial effects. Conclusions These findings support the rationale behind the encouragement of the development of drugs that repress the overexpression of FGFRs and suggest the dietary incorporation of supplementary nutrients in the management of degraded cartilage.
Collapse
Affiliation(s)
- Salvador Fernández-Arroyo
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, IISPV, Universitat Rovira i Virgili, Reus, Spain.,Campus of International Excellence Southern Catalonia, Tarragona, Spain;
| | - Fernando Huete-Toral
- Department of Biochemistry, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - María Jesús Pérez de Lara
- Department of Biochemistry, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - María de la Luz Cádiz-Gurrea
- Functional Food Research and Development Center, Health Science Technological Park, Granada, Spain.,Department of Analytical Chemistry, University of Granada, Granada, Spain
| | | | - Vicente Micol
- Institute of Molecular and Cell Biology, Miguel Hernández University, Elche, Spain
| | - Antonio Segura-Carretero
- Functional Food Research and Development Center, Health Science Technological Park, Granada, Spain.,Department of Analytical Chemistry, University of Granada, Granada, Spain
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, IISPV, Universitat Rovira i Virgili, Reus, Spain.,Campus of International Excellence Southern Catalonia, Tarragona, Spain
| | - Jesús Pintor
- Department of Biochemistry, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain;
| |
Collapse
|