1
|
Wen L, Liu Z, Zhou L, Liu Z, Li Q, Geng B, Xia Y. Bone and Extracellular Signal-Related Kinase 5 (ERK5). Biomolecules 2024; 14:556. [PMID: 38785963 PMCID: PMC11117709 DOI: 10.3390/biom14050556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/17/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Bones are vital for anchoring muscles, tendons, and ligaments, serving as a fundamental element of the human skeletal structure. However, our understanding of bone development mechanisms and the maintenance of bone homeostasis is still limited. Extracellular signal-related kinase 5 (ERK5), a recently identified member of the mitogen-activated protein kinase (MAPK) family, plays a critical role in the pathogenesis and progression of various diseases, especially neoplasms. Recent studies have highlighted ERK5's significant role in both bone development and bone-associated pathologies. This review offers a detailed examination of the latest research on ERK5 in different tissues and diseases, with a particular focus on its implications for bone health. It also examines therapeutic strategies and future research avenues targeting ERK5.
Collapse
Affiliation(s)
- Lei Wen
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming 650032, China
| | - Zirui Liu
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Libo Zhou
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Zhongcheng Liu
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Qingda Li
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Bin Geng
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Yayi Xia
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| |
Collapse
|
2
|
Liu P, Tu J, Wang W, Li Z, Li Y, Yu X, Zhang Z. Effects of Mechanical Stress Stimulation on Function and Expression Mechanism of Osteoblasts. Front Bioeng Biotechnol 2022; 10:830722. [PMID: 35252138 PMCID: PMC8893233 DOI: 10.3389/fbioe.2022.830722] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoclasts and osteoblasts play a major role in bone tissue homeostasis. The homeostasis and integrity of bone tissue are maintained by ensuring a balance between osteoclastic and osteogenic activities. The remodeling of bone tissue is a continuous ongoing process. Osteoclasts mainly play a role in bone resorption, whereas osteoblasts are mainly involved in bone remodeling processes, such as bone cell formation, mineralization, and secretion. These cell types balance and restrict each other to maintain bone tissue metabolism. Bone tissue is very sensitive to mechanical stress stimulation. Unloading and loading of mechanical stress are closely related to the differentiation and formation of osteoclasts and bone resorption function as well as the differentiation and formation of osteoblasts and bone formation function. Consequently, mechanical stress exerts an important influence on the bone microenvironment and bone metabolism. This review focuses on the effects of different forms of mechanical stress stimulation (including gravity, continuously compressive pressure, tensile strain, and fluid shear stress) on osteoclast and osteoblast function and expression mechanism. This article highlights the involvement of osteoclasts and osteoblasts in activating different mechanical transduction pathways and reports changings in their differentiation, formation, and functional mechanism induced by the application of different types of mechanical stress to bone tissue. This review could provide new ideas for further microscopic studies of bone health, disease, and tissue damage reconstruction.
Collapse
Affiliation(s)
- Pan Liu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Ji Tu
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Wenzhao Wang
- Department of Orthopedics, West China Hospital of Sichuan University, Chengdu, China
| | - Zheng Li
- People’s Hospital of Jiulongpo District, Chongqing, China
| | - Yao Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xiaoping Yu
- School of Public Health, Chengdu Medical College, Chengdu, China
- Basic Medical College of Chengdu University, Chengdu, China
- *Correspondence: Xiaoping Yu, ; Zhengdong Zhang,
| | - Zhengdong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- *Correspondence: Xiaoping Yu, ; Zhengdong Zhang,
| |
Collapse
|
3
|
Gould NR, Torre OM, Leser JM, Stains JP. The cytoskeleton and connected elements in bone cell mechano-transduction. Bone 2021; 149:115971. [PMID: 33892173 PMCID: PMC8217329 DOI: 10.1016/j.bone.2021.115971] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/30/2021] [Accepted: 04/17/2021] [Indexed: 02/07/2023]
Abstract
Bone is a mechano-responsive tissue that adapts to changes in its mechanical environment. Increases in strain lead to increased bone mass acquisition, whereas decreases in strain lead to a loss of bone mass. Given that mechanical stress is a regulator of bone mass and quality, it is important to understand how bone cells sense and transduce these mechanical cues into biological changes to identify druggable targets that can be exploited to restore bone cell mechano-sensitivity or to mimic mechanical load. Many studies have identified individual cytoskeletal components - microtubules, actin, and intermediate filaments - as mechano-sensors in bone. However, given the high interconnectedness and interaction between individual cytoskeletal components, and that they can assemble into multiple discreet cellular structures, it is likely that the cytoskeleton as a whole, rather than one specific component, is necessary for proper bone cell mechano-transduction. This review will examine the role of each cytoskeletal element in bone cell mechano-transduction and will present a unified view of how these elements interact and work together to create a mechano-sensor that is necessary to control bone formation following mechanical stress.
Collapse
Affiliation(s)
- Nicole R Gould
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Olivia M Torre
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jenna M Leser
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joseph P Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA..
| |
Collapse
|
4
|
Zhang B, An L, Geng B, Ding N, Coalson E, Wan L, Yan L, Mohammed FHA, Ma C, Li R, Yang X, Zhang X, Wang C, Ma J, Xia Y. ERK5 negatively regulates Kruppel-like factor 4 and promotes osteogenic lineage cell proliferation in response to MEK5 overexpression or fluid shear stress. Connect Tissue Res 2021; 62:194-205. [PMID: 31749391 DOI: 10.1080/03008207.2019.1670650] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Aim of the study: Fluid shear stress (FSS) plays a critical role in osteoblast proliferation via extracellular signal-regulated kinase 5 (ERK5). Kruppel-like factor 4 (KLF4) knockout robustly enhances bone formation due to increased osteoblast differentiation and mineralization. However, the effect of KLF4 on osteoblast proliferation is unresolved. Therefore, the aim of our study was to investigate the effect of KLF4 on osteogenic lineage cell proliferation and the relationship between KLF4 and ERK5. Materials and methods: MC3T3-E1 cells were treated with FSS and/or KLF4 siRNA, cell viability was accessed by Edu labeling and CCK-8 assay, and proliferative gene expression were assessed by PCR array. Bone marrow stromal cells (BMSCs) were infected with adenovirus expressing KLF4 and/or constitutively active MEK5, cell viability was evaluated using crystal violet staining, colony formation assay, and cell WST1 assay. The levels of KLF4 and ERK5 phosphorylation were identified through qRT-PCR and western blot, respectively. Results: KLF4 expression was significantly down-regulated by FSS exposure, however, this was reversed by ERK5 siRNA. KLF4 overexpression inhibited colony formation efficiency and cell viability in BMSCs. Adenoviruses expressing constitutively active MEK5 increased ERK5 phosphorylation, which inhibited KLF4 expression, and promoted BMSC proliferation. FSS-induced osteoblast proliferation also involved elevation of Cyclin B2 and Cdc14b as well as repressed expression of P27. Conclusions: KLF4 negatively regulates osteogenic lineage cell proliferation, and ERK5 negatively regulates KLF4 expression and promotes osteogenic lineage cell proliferation.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Liping An
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Bin Geng
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Ning Ding
- Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, People's Hospital of Gansu Province , Lanzhou, Gansu, China
| | - Elam Coalson
- Pritzker School of Medicine, University of Chicago , Chicago, IL, USA
| | - Lang Wan
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Liang Yan
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Fawaz H A Mohammed
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Chongwen Ma
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Rui Li
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Xinxin Yang
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Xiaohui Zhang
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Cuifang Wang
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Jinglin Ma
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China.,Department of Orthopaedic Surgery, Key Laboratory of Orthopedics of Gansu Province , Lanzhou, Gansu, China
| | - Yayi Xia
- Department of Orthopaedic Surgery or Institute of Bone and Joint Research, The Second Hospital of Lanzhou University , Lanzhou, Gansu, China
| |
Collapse
|
5
|
Ma C, Geng B, Zhang X, Li R, Yang X, Xia Y. Fluid Shear Stress Suppresses Osteoclast Differentiation in RAW264.7 Cells through Extracellular Signal-Regulated Kinase 5 (ERK5) Signaling Pathway. Med Sci Monit 2020; 26:e918370. [PMID: 31914120 PMCID: PMC6977602 DOI: 10.12659/msm.918370] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Although extracellular signal-regulated kinase 5 (ERK5) is known to be critical for osteoclast differentiation, there are few studies on how fluid shear stress (FSS) regulates osteoclast differentiation through the ERK5 signaling pathway. We examined the expression of nuclear factor of activated T cells c1 (NFATc1) in RAW264.7 cells and its downstream factors, including cathepsin K (CTSK), tartrate-resistant acid phosphatase (TRAP), matrix metalloproteinases-9 (MMP-9) and their relationship with ERK5. Material/Methods RAW264.7 cells were treated with RANKL, XMD8-92 (ERK5 inhibitor), and then loaded onto 12 dyn/cm2 FSS for 4 days. Endpoints measured were osteoclast differentiation, bone resorption, and TRAP activity. Cell viability was detected by using the Cell Counting Kit-8 (CCK-8) assay. Western blot was used to analyze protein expression of phosphorylated-ERK5 (p-ERK5), NFATc1, CTSK, TRAP, and MMP-9. Results FSS inhibited osteoclast differentiation and expression of NFATc1, CTSK, TRAP, and MMP-9; cell viability was not affected. ERK5 expression increased by FSS but not by RANKL, and it was blocked by XMD8-92. Furthermore, FSS suppressed osteoclast differentiation in RAW264.7 cells through ERK5 pathway. Conclusions Our findings demonstrated that FSS inhibited osteoclast differentiation in RAW264.7 cells via the ERK5 pathway through reduced NFATc1 expression and its downstream factors MMP-9, CTSK, and TRAP.
Collapse
Affiliation(s)
- Chongwen Ma
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| | - Bin Geng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| | - Xiaohui Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| | - Rui Li
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| | - Xinxin Yang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| | - Yayi Xia
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China (mainland).,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China (mainland)
| |
Collapse
|
6
|
Ding N, Geng B, Li Z, Yang Q, Yan L, Wan L, Zhang B, Wang C, Xia Y. Fluid shear stress promotes osteoblast proliferation through the NFATc1-ERK5 pathway. Connect Tissue Res 2019; 60:107-116. [PMID: 29609502 DOI: 10.1080/03008207.2018.1459588] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Extracellular-regulated kinase 5 (ERK5) is thought to regulate osteoblast proliferation. To further understand how ERK5 signaling regulates osteoblast proliferation induced by fluid shear stress (FSS), we examined some potential signaling targets associated with ERK5 in MC3T3-E1 cells. METHODS MC3T3-E1 cells were treated with XMD8-92 (an ERK5 inhibitor) or Cyclosporin A (CsA, a nuclear factor of activated T cells (NFAT) c1 inhibitor) and/or exposed to 12 dyn/cm2 FSS. Phosphorylated-ERK5 (p-ERK5) and expression levels of NFATc1, ERK5, E2F2, and cyclin E1 were analyzed by western blot. The mRNA levels of genes associated with cell proliferation were analyzed by Polymerase Chain Reaction (PCR) array. Subcellular localization of p-ERK5 and NFATc1 were determined by immunofluorescence. Cell proliferation was evaluated by MTT assay. RESULTS NFATc1 expression was up-regulated by FSS. XMD8-92 only blocked ERK5 activation; however, CsA decreased NFATc1 and p-ERK5 levels, including after FSS stimulation. Exposure to NFATc1 inhibitor or ERK5 inhibitor resulted in decreased E2F2 and cyclin E1 expression and proliferation by proliferative MC3T3-E1 cells. Furthermore, immunofluorescence results illustrated that NFATc1 induced ERK5 phosphorylation, resulting in p-ERK5 translocation to the nucleus. CONCLUSIONS Our results reveal that NFATc1 acts as an intermediate to promote the phosphorylation of ERK5 induced by FSS. Moreover, activated NFATc1-ERK5 signaling up-regulates the expression of E2F2 and cyclin E1, which promote osteoblast proliferation.
Collapse
Affiliation(s)
- Ning Ding
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Bin Geng
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Zhonghao Li
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Quanzeng Yang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Liang Yan
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Lang Wan
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Bo Zhang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Cuifang Wang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Yayi Xia
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| |
Collapse
|
7
|
Shuai C, Yang W, Peng S, Gao C, Guo W, Lai Y, Feng P. Physical stimulations and their osteogenesis-inducing mechanisms. Int J Bioprint 2018; 4:138. [PMID: 33102916 PMCID: PMC7581999 DOI: 10.18063/ijb.v4i2.138] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/09/2018] [Indexed: 12/27/2022] Open
Abstract
Physical stimulations such as magnetic, electric and mechanical stimulation could enhance cell activity and promote bone formation in bone repair process via activating signal pathways, modulating ion channels, regulating bonerelated gene expressions, etc. In this paper, bioeffects of physical stimulations on cell activity, tissue growth and bone healing were systematically summarized, which especially focused on their osteogenesis-inducing mechanisms. Detailedly, magnetic stimulation could produce Hall effect which improved the permeability of cell membrane and promoted the migration of ions, especially accelerating the extracellular calcium ions to pass through cell membrane. Electric stimulation could induce inverse piezoelectric effect which generated electric signals, accordingly up-regulating intracellular calcium levels and growth factor synthesis. And mechanical stimulation could produce mechanical signals which were converted into corresponding biochemical signals, thus activating various signaling pathways on cell membrane and inducing a series of gene expressions. Besides, bioeffects of physical stimulations combined with bone scaffolds which fabricated using 3D printing technology on bone cells were discussed. The equipments of physical stimulation system were described. The opportunities and challenges of physical stimulations were also presented from the perspective of bone repair.
Collapse
Affiliation(s)
- Cijun Shuai
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China.,Jiangxi University of Science and Technology, Ganzhou, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Wenjing Yang
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Shuping Peng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Chengde Gao
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Wang Guo
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Yuxiao Lai
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| | - Pei Feng
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| |
Collapse
|
8
|
Fluid flow facilitates inward rectifier K + current by convectively restoring [K +] at the cell membrane surface. Sci Rep 2016; 6:39585. [PMID: 28004830 PMCID: PMC5177964 DOI: 10.1038/srep39585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 11/24/2016] [Indexed: 01/25/2023] Open
Abstract
The inward rectifier Kir2.1 current (IKir2.1) was reported to be facilitated by fluid flow. However, the mechanism underlying this facilitation remains uncertain. We hypothesized that during K+ influx or efflux, [K+] adjacent to the outer mouth of the Kir2.1 channel might decrease or increase, respectively, compared with the average [K+] of the bulk extracellular solution, and that fluid flow could restore the original [K+] and result in the apparent facilitation of IKir2.1. We recorded the IKir2.1 in RBL-2H3 cells and HEK293T cells that were ectopically over-expressed with Kir2.1 channels by using the whole-cell patch-clamp technique. Fluid-flow application immediately increased the IKir2.1, which was not prevented by either the pretreatment with inhibitors of various protein kinases or the modulation of the cytoskeleton and caveolae. The magnitudes of the increases of IKir2.1 by fluid flow were driving force-dependent. Simulations performed using the Nernst-Planck mass equation indicated that [K+] near the membrane surface fell markedly below the average [K+] of the bulk extracellular solution during K+ influx, and, notably, that fluid flow restored the decreased [K+] at the cell surface in a flow rate-dependent manner. These results support the “convection-regulation hypothesis” and define a novel interpretation of fluid flow-induced modulation of ion channels.
Collapse
|
9
|
Bo Z, Bin G, Jing W, Cuifang W, Liping A, Jinglin M, Jin J, Xiaoyi T, Cong C, Ning D, Yayi X. Fluid shear stress promotes osteoblast proliferation via the Gαq-ERK5 signaling pathway. Connect Tissue Res 2016; 57:299-306. [PMID: 27115838 DOI: 10.1080/03008207.2016.1181063] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Fluid shear stress (FSS) is a ubiquitous mechanical stimulus that potently promotes osteoblast proliferation. Previously, we reported that extracellular signal-regulated kinase 5 (ERK5) is essential for FSS-induced osteoblast proliferation. However, the precise mechanism by which FSS promotes osteoblast proliferation via ERK5 activation is poorly understood. The aim of this study was to determine the critical role of Gαq in FSS-induced ERK5 phosphorylation and osteoblast proliferation, as well as the downstream targets of the Gαq-ERK5 pathway. MC3T3-E1 cells were transfected with 50 nM Gαq siRNA, treated with 5 mM XMD8-92 (a highly selective inhibitor of ERK5 activity), and/or exposed to FSS (12 dyn/cm(2)). Cell proliferation was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The protein expression levels of Gαq, P-ERK5, ERK5, Cyclin B1, and CDK1 were analyzed by Western blot. Physiological FSS exposure for 60 min remarkably promoted MC3T3-E1 cell proliferation, however, this effect was suppressed by siRNA-mediated Gαq knockdown or inhibition of ERK5 activity by XMD8-92 treatment, suggesting that Gαq and ERK5 might modulate FSS-increased osteoblast proliferation. Furthermore, ERK5 phosphorylation was dramatically inhibited by Gαq siRNA. In addition, our study further revealed that FSS treatment of MC3T3-E1 cells for 60 min markedly upregulated the protein expression levels of Cyclin B1 and CDK1, and this increased expression was predominantly blocked by Gαq siRNA or XMD8-92 treatment. We propose that FSS acts on the Gαq-ERK5 signaling pathway to upregulate Cyclin B1 and CDK1 expression, thereby resulting in MC3T3-E1 cell proliferation. Thus, the Gαq-ERK5 signaling pathway may provide useful information regarding the treatment of bone metabolic disease.
Collapse
Affiliation(s)
- Zhang Bo
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Geng Bin
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Wang Jing
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Wang Cuifang
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - An Liping
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Ma Jinglin
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Jiang Jin
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Tan Xiaoyi
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Chen Cong
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Ding Ning
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Xia Yayi
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| |
Collapse
|
10
|
Bin G, Bo Z, Jing W, Jin J, Xiaoyi T, Cong C, Liping A, Jinglin M, Cuifang W, Yonggang C, Yayi X. Fluid shear stress suppresses TNF-α-induced apoptosis in MC3T3-E1 cells: Involvement of ERK5-AKT-FoxO3a-Bim/FasL signaling pathways. Exp Cell Res 2016; 343:208-217. [PMID: 27060196 DOI: 10.1016/j.yexcr.2016.03.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 03/08/2016] [Accepted: 03/16/2016] [Indexed: 10/22/2022]
Abstract
TNF-α is known to induce osteoblasts apoptosis, whereas mechanical stimulation has been shown to enhance osteoblast survival. In the present study, we found that mechanical stimulation in the form of fluid shear stress (FSS) suppresses TNF-α induced apoptosis in MC3T3-E1 cells. Extracellular signal-regulated kinase 5 (ERK5) is a member of the mitogen-activated protein kinase (MAPK) family that has been implicated in cell survival. We also demonstrated that FSS imposed by flow chamber in vitro leads to a markedly activation of ERK5, which was shown to be protective against TNF-α-induced apoptosis, whereas the transfection of siRNA against ERK5 (ERK5-siRNA) reversed the FSS-medicated anti-apoptotic effects. An initial FSS-mediated activation of ERK5 that phosphorylates AKT to increase its activity, and a following forkhead box O 3a (FoxO3a) was phosphorylated by activated AKT. Phosphorylated FoxO3a is sequestered in the cytoplasm, and prevents it from translocating to nucleus where it can increase the expression of FasL and Bim. The inhibition of AKT-FoxO3a signalings by a PI3K (PI3-kinase)/AKT inhibitor (LY294002) or the transfection of ERK5-siRNA led to the nuclear translocation of non-phosphorylated FoxO3a, and increased the protein expression of FasL and Bim. In addition, the activation of caspase-3 by TNF-α was significantly inhibited by aforementioned FSS-medicated mechanisms. In brief, the activation of ERK5-AKT-FoxO3a signaling pathways by FSS resulted in a decreased expression of FasL and Bim and an inhibition of caspase-3 activation, which exerts a protective effect that prevents osteoblasts from apoptosis.
Collapse
Affiliation(s)
- Geng Bin
- The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, 730000 Gansu, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000 Gansu, China
| | - Zhang Bo
- The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, 730000 Gansu, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000 Gansu, China
| | - Wang Jing
- The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, 730000 Gansu, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000 Gansu, China
| | - Jiang Jin
- The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, 730000 Gansu, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000 Gansu, China
| | - Tan Xiaoyi
- The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, 730000 Gansu, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000 Gansu, China
| | - Chen Cong
- The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, 730000 Gansu, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000 Gansu, China
| | - An Liping
- The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, 730000 Gansu, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000 Gansu, China
| | - Ma Jinglin
- The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, 730000 Gansu, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000 Gansu, China
| | - Wang Cuifang
- The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, 730000 Gansu, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000 Gansu, China
| | - Chen Yonggang
- The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, 730000 Gansu, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000 Gansu, China
| | - Xia Yayi
- The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, 730000 Gansu, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000 Gansu, China.
| |
Collapse
|
11
|
Bin G, Cuifang W, Bo Z, Jing W, Jin J, Xiaoyi T, Cong C, Yonggang C, Liping A, Jinglin M, Yayi X. Fluid shear stress inhibits TNF-α-induced osteoblast apoptosis via ERK5 signaling pathway. Biochem Biophys Res Commun 2015; 466:117-23. [DOI: 10.1016/j.bbrc.2015.08.117] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 08/26/2015] [Indexed: 02/06/2023]
|
12
|
Jiang J, Zhao LG, Teng YJ, Chen SL, An LP, Ma JL, Wang J, Xia YY. ERK5 signalling pathway is essential for fluid shear stress-induced COX-2 gene expression in MC3T3-E1 osteoblast. Mol Cell Biochem 2015; 406:237-43. [DOI: 10.1007/s11010-015-2441-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 05/06/2015] [Indexed: 12/20/2022]
|
13
|
Tagliaferri C, Wittrant Y, Davicco MJ, Walrand S, Coxam V. Muscle and bone, two interconnected tissues. Ageing Res Rev 2015; 21:55-70. [PMID: 25804855 DOI: 10.1016/j.arr.2015.03.002] [Citation(s) in RCA: 248] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/15/2015] [Accepted: 03/18/2015] [Indexed: 12/31/2022]
Abstract
As bones are levers for skeletal muscle to exert forces, both are complementary and essential for locomotion and individual autonomy. In the past decades, the idea of a bone-muscle unit has emerged. Numerous studies have confirmed this hypothesis from in utero to aging works. Space flight, bed rest as well as osteoporosis and sarcopenia experimentations have allowed to accumulate considerable evidence. Mechanical loading is a key mechanism linking both tissues with a central promoting role of physical activity. Moreover, the skeletal muscle secretome accounts various molecules that affect bone including insulin-like growth factor-1 (IGF-1), basic fibroblast growth factor (FGF-2), interleukin-6 (IL-6), IL-15, myostatin, osteoglycin (OGN), FAM5C, Tmem119 and osteoactivin. Even though studies on the potential effects of bone on muscle metabolism are sparse, few osteokines have been identified. Prostaglandin E2 (PGE2) and Wnt3a, which are secreted by osteocytes, osteocalcin (OCN) and IGF-1, which are produced by osteoblasts and sclerostin which is secreted by both cell types, might impact skeletal muscle cells. Cartilage and adipose tissue are also likely to participate to this control loop and should not be set aside. Indeed, chondrocytes are known to secrete Dickkopf-1 (DKK-1) and Indian hedgehog (Ihh) and adipocytes produce leptin, adiponectin and IL-6, which potentially modulate bone and muscle metabolisms. The understanding of this system will enable to define new levers to prevent/treat sarcopenia and osteoporosis at the same time. These strategies might include nutritional interventions and physical exercise.
Collapse
|
14
|
Carrisoza-Gaytan R, Liu Y, Flores D, Else C, Lee HG, Rhodes G, Sandoval RM, Kleyman TR, Lee FYI, Molitoris B, Satlin LM, Rohatgi R. Effects of biomechanical forces on signaling in the cortical collecting duct (CCD). Am J Physiol Renal Physiol 2014; 307:F195-204. [PMID: 24872319 DOI: 10.1152/ajprenal.00634.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
An increase in tubular fluid flow rate (TFF) stimulates Na reabsorption and K secretion in the cortical collecting duct (CCD) and subjects cells therein to biomechanical forces including fluid shear stress (FSS) and circumferential stretch (CS). Intracellular MAPK and extracellular autocrine/paracrine PGE2 signaling regulate cation transport in the CCD and, at least in other systems, are affected by biomechanical forces. We hypothesized that FSS and CS differentially affect MAPK signaling and PGE2 release to modulate cation transport in the CCD. To validate that CS is a physiological force in vivo, we applied the intravital microscopic approach to rodent kidneys in vivo to show that saline or furosemide injection led to a 46.5 ± 2.0 or 170 ± 32% increase, respectively, in distal tubular diameter. Next, murine CCD (mpkCCD) cells were grown on glass or silicone coated with collagen type IV and subjected to 0 or 0.4 dyne/cm(2) of FSS or 10% CS, respectively, forces chosen based on prior biomechanical modeling of ex vivo microperfused CCDs. Cells exposed to FSS expressed an approximately twofold greater abundance of phospho(p)-ERK and p-p38 vs. static cells, while CS did not alter p-p38 and p-ERK expression compared with unstretched controls. FSS induced whereas CS reduced PGE2 release by ∼40%. In conclusion, FSS and CS differentially affect ERK and p38 activation and PGE2 release in a cell culture model of the CD. We speculate that TFF differentially regulates biomechanical signaling and, in turn, cation transport in the CCD.
Collapse
Affiliation(s)
| | - Yu Liu
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Daniel Flores
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Medicine, James J. Peters Veterans Affairs Medical Center, New York, New York
| | - Cindy Else
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Heon Goo Lee
- Department of Orthopedics, Robert Carroll and Jane Chace Carroll Laboratories, Columbia College of Physicians and Surgeons, New York, New York
| | - George Rhodes
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Ruben M Sandoval
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Thomas R Kleyman
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Francis Young-In Lee
- Department of Orthopedics, Robert Carroll and Jane Chace Carroll Laboratories, Columbia College of Physicians and Surgeons, New York, New York
| | - Bruce Molitoris
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Lisa M Satlin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rajeev Rohatgi
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Medicine, James J. Peters Veterans Affairs Medical Center, New York, New York;
| |
Collapse
|
15
|
Zhao LG, Chen SL, Teng YJ, An LP, Wang J, Ma JL, Xia YY. The MEK5/ERK5 pathway mediates fluid shear stress promoted osteoblast differentiation. Connect Tissue Res 2014; 55:96-102. [PMID: 24111522 DOI: 10.3109/03008207.2013.853755] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The aim of this study was to determine the role of the mitogen-activated protein kinase kinase (MEK) 5/extracellular signal-regulated kinase (ERK) 5 pathway in osteoblast differentiation promoted by intermittent fluid shear stress (FSS). MC3T3-E1 osteoblastic cells were subjected to 12 dyn/cm(2) intermittent FSS, and the phenotypic markers for osteoblast differentiation, such as alkaline phosphatase (ALP) activity and expression of osteopontin (OPN) and osteocalcin (OCN), were then examined. The results showed that intermittent FSS could stimulate ERK5 phosphorylation, ALP activity and the expression of OPN and OCN. When the MEK5/ERK5 pathway was selectively inhibited by BIX02189, ALP activity was suppressed, and the expression of OPN and OCN was downregulated. Intermittent FSS induce the expression of Runt-related transcription factor-2 (Runx-2), which is involved in osteoblast differentiation by promoting the transcription of the above genes. Furthermore, the expression of Runx-2 was also reduced after treatment with BIX02189. Finally, we found that intermittent FSS was a more intense stimulus than steady FSS for promoting osteoblast differentiation. In summary, our results suggest that the MEK5/ERK5 pathway mediates osteoblast differentiation promoted by intermittent FSS, which was more effective than steady FSS in the differentiation process. The MEK5/ERK5 pathway also mediates FSS-induced Runx-2 expression in osteoblast differentiation.
Collapse
Affiliation(s)
- Liang-gong Zhao
- The Second Clinical Medical College of Lanzhou University , Lanzhou, Gansu Province , P.R. China
| | | | | | | | | | | | | |
Collapse
|
16
|
Yang HY, Kwon J, Kook MS, Kang SS, Kim SE, Sohn S, Jung S, Kwon SO, Kim HS, Lee JH, Lee TH. Proteomic analysis of gingival tissue and alveolar bone during alveolar bone healing. Mol Cell Proteomics 2013; 12:2674-88. [PMID: 23824910 DOI: 10.1074/mcp.m112.026740] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Bone tissue regeneration is orchestrated by the surrounding supporting tissues and involves the build-up of osteogenic cells, which orchestrate remodeling/healing through the expression of numerous mediators and signaling molecules. Periodontal regeneration models have proven useful for studying the interaction and communication between alveolar bone and supporting soft tissue. We applied a quantitative proteomic approach to analyze and compare proteins with altered expression in gingival soft tissue and alveolar bone following tooth extraction. For target identification and validation, hard and soft tissue were extracted from mini-pigs at the indicated times after tooth extraction. From triplicate experiments, 56 proteins in soft tissue and 27 proteins in alveolar bone were found to be differentially expressed before and after tooth extraction. The expression of 21 of those proteins was altered in both soft tissue and bone. Comparison of the activated networks in soft tissue and alveolar bone highlighted their distinct responsibilities in bone and tissue healing. Moreover, we found that there is crosstalk between identified proteins in soft tissue and alveolar bone with respect to cellular assembly, organization, and communication. Among these proteins, we examined in detail the expression patterns and associated networks of ATP5B and fibronectin 1. ATP5B is involved in nucleic acid metabolism, small molecule biochemistry, and neurological disease, and fibronectin 1 is involved in cellular assembly, organization, and maintenance. Collectively, our findings indicate that bone regeneration is accompanied by a profound interaction among networks regulating cellular resources, and they provide novel insight into the molecular mechanisms involved in the healing of periodontal tissue after tooth extraction.
Collapse
Affiliation(s)
- Hee-Young Yang
- Department of Oral Biochemistry, Dental Science Research Institute and the BK21 Project, Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Peng Q, Qiu J, Sun J, Yang L, Zhang B, Wang Y. The nuclear localization of MGF receptor in osteoblasts under mechanical stimulation. Mol Cell Biochem 2012; 369:147-56. [PMID: 22752413 DOI: 10.1007/s11010-012-1377-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 06/20/2012] [Indexed: 12/13/2022]
Abstract
Mechano-growth factor (MGF) has emerged as an important mechanosensitive player in bone repair, but understanding of MGF function is hampered by the fact that MGF receptor and the underlying pathways remain unknown. In this study, fluorescein isothiocyanate (FITC)-labeled MGF-Ct24E (FITC-MGF) was used to determine the subcellular localization of MGF receptor in osteoblasts. After the primary osteoblasts were exposed to stretch with the strain at 10 %, and/or loaded with 50 ng/ml exogenous MGF-Ct24E, cells were incubated with the different concentrations of FITC-MGF (0.01, 0.1, and 1 mg/ml) followed by flow cytometry and laser scanning confocal microscope analysis. Our results showed that the fluorescence intensity and cell population internalizing FITC-MGF increased with the concentration of FITC-MGF. And all the cells were labeled with fluorescence at 1 mg/ml. Notably, FITC-MGF had nuclear localization when osteoblasts were exposed to stretch and/or 50 ng/ml MGF-Ct24E added, compared to the evident cytoplasmic localization in the static culture group. The nuclear localization of FITC-MGF in response to mechanical loading was found to associate with high expression of proliferating cell nuclear antigen, suggesting MGF and its receptor could serve as potential messengers that replay information in nuclei to control cell proliferation.
Collapse
Affiliation(s)
- Qin Peng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, People's Republic of China
| | | | | | | | | | | |
Collapse
|