1
|
Yin C, Deng M, Yu J, Chen Y, Zheng K, Huang Y, Deng X, Tian Y, Ma Y, Zeng B, Guo X, Guo B. An Andrias davidianus derived composite hydrogel with enhanced antibacterial and bone repair properties for osteomyelitis treatment. Sci Rep 2024; 14:24626. [PMID: 39426986 PMCID: PMC11490572 DOI: 10.1038/s41598-024-75997-8] [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: 05/02/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
Effective antibacterial therapy while accelerating the repair of bone defects is crucial for the treatment of osteomyelitis. Inspired by the protective mechanism of Andrias davidianus, we constructed an antibacterial hydrogel scaffold with excellent rigidity and long-term slow-release activity. While retaining the toughness of the skin secretion of Andrias davidianus (SSAD), the rigidity of the hydrogel material is increased by incorporating hydroxyapatite to meet the demands of bone-defect-filling materials. It also exerted antibacterial effects via the slow-release of vancomycin from local osteomyelitis lesions. Notably, the hydrogel can also carry a high stable recombinant miR-214-3p inhibitor (MSA-anti214). By the delivery of nano vector polyvinylamine, the long-term slow-release of MSA-anti214 is achieved to promote bone repair, making this composite hydrogel a potential SSAD-based osteomyelitis alleviator (SOA). In vitro and vivo results verified that the SOA effectively eliminated Staphylococcus aureus and repaired bone defects, ultimately mitigating the progression of osteomyelitis. This composite hydrogel extends the economic application prospects of A. davidianus and has provided new insights for the treatment of osteomyelitis. The study also explored new insights for the bone filling materials of bone defection and other skeletal system diseases.
Collapse
Affiliation(s)
- Chong Yin
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Meng Deng
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Jinshu Yu
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Yonghao Chen
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Kaiyuan Zheng
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Yi Huang
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Xudong Deng
- Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Ye Tian
- Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Yuwen Ma
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Beilei Zeng
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Xiaolan Guo
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
| | - Bin Guo
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
- School of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
| |
Collapse
|
2
|
Hu L, Chen W, Qian A, Li YP. Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and disease. Bone Res 2024; 12:39. [PMID: 38987555 PMCID: PMC11237130 DOI: 10.1038/s41413-024-00342-8] [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: 12/10/2023] [Revised: 04/27/2024] [Accepted: 05/12/2024] [Indexed: 07/12/2024] Open
Abstract
Wnts are secreted, lipid-modified proteins that bind to different receptors on the cell surface to activate canonical or non-canonical Wnt signaling pathways, which control various biological processes throughout embryonic development and adult life. Aberrant Wnt signaling pathway underlies a wide range of human disease pathogeneses. In this review, we provide an update of Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and diseases. The Wnt proteins, receptors, activators, inhibitors, and the crosstalk of Wnt signaling pathways with other signaling pathways are summarized and discussed. We mainly review Wnt signaling functions in bone formation, homeostasis, and related diseases, and summarize mouse models carrying genetic modifications of Wnt signaling components. Moreover, the therapeutic strategies for treating bone diseases by targeting Wnt signaling, including the extracellular molecules, cytosol components, and nuclear components of Wnt signaling are reviewed. In summary, this paper reviews our current understanding of the mechanisms by which Wnt signaling regulates bone formation, homeostasis, and the efforts targeting Wnt signaling for treating bone diseases. Finally, the paper evaluates the important questions in Wnt signaling to be further explored based on the progress of new biological analytical technologies.
Collapse
Affiliation(s)
- Lifang Hu
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Airong Qian
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
3
|
Salem D, Fecek RJ. Role of microtubule actin crosslinking factor 1 (MACF1) in bipolar disorder pathophysiology and potential in lithium therapeutic mechanism. Transl Psychiatry 2023; 13:221. [PMID: 37353479 DOI: 10.1038/s41398-023-02483-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/05/2023] [Accepted: 05/23/2023] [Indexed: 06/25/2023] Open
Abstract
Bipolar affective disorder (BPAD) are life-long disorders that account for significant morbidity in afflicted patients. The etiology of BPAD is complex, combining genetic and environmental factors to increase the risk of disease. Genetic studies have pointed toward cytoskeletal dysfunction as a potential molecular mechanism through which BPAD may arise and have implicated proteins that regulate the cytoskeleton as risk factors. Microtubule actin crosslinking factor 1 (MACF1) is a giant cytoskeletal crosslinking protein that can coordinate the different aspects of the mammalian cytoskeleton with a wide variety of actions. In this review, we seek to highlight the functions of MACF1 in the nervous system and the molecular mechanisms leading to BPAD pathogenesis. We also offer a brief perspective on MACF1 and the role it may be playing in lithium's mechanism of action in treating BPAD.
Collapse
Affiliation(s)
- Deepak Salem
- Lake Erie College of Osteopathic Medicine at Seton Hill, Department of Microbiology, Greensburg, USA
- University of Maryland Medical Center/Sheppard Pratt Psychiatry Residency Program, Baltimore, USA
| | - Ronald J Fecek
- Lake Erie College of Osteopathic Medicine at Seton Hill, Department of Microbiology, Greensburg, USA.
| |
Collapse
|
4
|
Wang P, Zhang J, Zhang H, Zhang F. The role of MACF1 on acute myeloid leukemia cell proliferation is involved in Runx2-targeted PI3K/Akt signaling. Mol Cell Biochem 2023; 478:433-441. [PMID: 35857251 DOI: 10.1007/s11010-022-04517-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 06/24/2022] [Indexed: 11/25/2022]
Abstract
Acute myeloid leukemia (AML) is a type of hematologic diseases, which is related to abnormal genes. The aberrant microtubule actin cross-linking factor 1 (MACF1) is associated with progression of multiple tumors by initiating cell proliferation. Nevertheless, the function and action mechanism of MACF1 in AML cell proliferation remain mostly unknown. Our study aimed to explore the influence of MACF1 on AML cell proliferation by CCK-8 and EdU staining assays. Moreover, we aimed to explore the effect of MACF1 on downstream Runx2 and the PI3K/Akt signaling. MACF1 expression in AML patients was predicted by bioinformatics analysis. Cells were transfected with si-con, si-MACF1 or Runx2 using Lipofectamine 2000. Upregulated MACF1 was found in AML patients and predicted worse overall survival. MACF1 expression was upregulated in AML cells compared with that in hematopoietic stem and progenitor cells. MACF1 silencing reduced AML cell proliferation. Runx2 level was increased in AML cells, and decreased by silencing MACF1. Runx2 upregulation rescued MACF1 silencing-mediated inhibition of proliferation. MACF1 downregulation inhibited activation of the PI3K/Akt pathway by decreasing Runx2. Activation of the PI3K/Akt pathway abrogated the suppressive role of MACF1 downregulation in AML cell proliferation. In conclusion, MACF1 knockdown decreased AML cell proliferation by reducing Runx2 and inactivating the PI3K/Akt signaling.
Collapse
Affiliation(s)
- Ping Wang
- Department of Hematology, People's Hospital of Chongqing Banan District (Banan Hospital of Chongqing Medical University), No.659, Yu'nan Avenue, Chongqing, 401320, People's Republic of China.
| | - Jiajia Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Henan Polytechnic University, Jiaozuo, 454000, Henan, People's Republic of China
| | - Hui Zhang
- Department of Endocrinology, The First Affiliated Hospital of Henan Polytechnic University, Jiaozuo, 454000, Henan, People's Republic of China
| | - Fang Zhang
- Department of Neurology, The First Affiliated Hospital of Henan Polytechnic University, Jiaozuo, 454000, Henan, People's Republic of China
| |
Collapse
|
5
|
Jiang S, Yin C, Dang K, Zhang W, Huai Y, Qian A. Comprehensive ceRNA network for MACF1 regulates osteoblast proliferation. BMC Genomics 2022; 23:695. [PMID: 36207684 PMCID: PMC9541005 DOI: 10.1186/s12864-022-08910-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Previous studies have shown that microtubule actin crosslinking factor 1 (MACF1) can regulate osteoblast proliferation and differentiation through non-coding RNA (ncRNA) in bone-forming osteoblasts. However, the role of MACF1 in targeting the competing endogenous RNA (ceRNA) network to regulate osteoblast differentiation remains poorly understood. Here, we profiled messenger RNA (mRNA), microRNA (miRNA), and long ncRNA (lncRNA) expression in MACF1 knockdown MC3TC‑E1 pre‑osteoblast cells. RESULTS In total, 547 lncRNAs, 107 miRNAs, and 376 mRNAs were differentially expressed. Significantly altered lncRNAs, miRNAs, and mRNAs were primarily found on chromosome 2. A lncRNA-miRNA-mRNA network was constructed using a bioinformatics computational approach. The network indicated that mir-7063 and mir-7646 were the most potent ncRNA regulators and mef2c was the most potent target gene. Pathway enrichment analysis showed that the fluid shear stress and atherosclerosis, p53 signaling, and focal adhesion pathways were highly enriched and contributed to osteoblast proliferation. Importantly, the fluid shear stress and atherosclerosis pathway was co-regulated by lncRNAs and miRNAs. In this pathway, Dusp1 was regulated by AK079370, while Arhgef2 was regulated by mir-5101. Furthermore, Map3k5 was regulated by AK154638 and mir-466q simultaneously. AK003142 and mir-3082-5p as well as Ak141402 and mir-446 m-3p were identified as interacting pairs that regulate target genes. CONCLUSION This study revealed the global expression profile of ceRNAs involved in the differentiation of MC3TC‑E1 osteoblasts induced by MACF1 deletion. These results indicate that loss of MACF1 activates a comprehensive ceRNA network to regulate osteoblast proliferation.
Collapse
Affiliation(s)
- Shanfeng Jiang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Chong Yin
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Department of Clinical Laboratory, Academician (expert) workstation, Lab of epigenetics and RNA therapy, Affiliated Hospital of North Sichuan Medical College, 637000, Nanchong, China
| | - Kai Dang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Wenjuan Zhang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Ying Huai
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Airong Qian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China.
| |
Collapse
|
6
|
Su P, Tian Y, Yin C, Wang X, Li D, Yang C, Pei J, Deng X, King S, Li Y, Qian A. MACF1 promotes osteoblastic cell migration by regulating MAP1B through the GSK3beta/TCF7 pathway. Bone 2022; 154:116238. [PMID: 34700040 DOI: 10.1016/j.bone.2021.116238] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 11/29/2022]
Abstract
RATIONALE The migration of osteoblastic cells to bone formation surface is an essential step for bone development and growth. However, whether the migration capacity of osteoblastic cells is compromised during osteoporosis occurrence and how it contributes to bone formation reduction remain unexplored so far. In this work, we found, as a positive regulator of cell migration, microtubule actin crosslinking factor 1 (MACF1) enhanced osteoblastic cells migration. We also examined whether MACF1 could facilitate osteoblastic cells' migration to bone formation surface to promote bone formation through another cytoskeleton protein, microtubule associated protein 1 (MAP1B). METHODS Preosteoblast cell line MC3T3-E1 with different MACF1 level was used for in vitro and in vivo cell migration assay; Primary cortical bone derived mesenchymal stem cells (C-MSCs) from bone tissue of MACF1 conditional knock out (cKO) mice was used for in vitro cell migration assay. Cell migration ability in vitro was evaluated by wound healing assay and transwell assay and in vivo by bone marrow cavity injection. Small interfering RNA (siRNA) was used for knocking down Map1b in MC3T3-E1 cell. Lithium chloride (LiCl) and Wortmannin (Wort) were used for inhibiting/activating GSK3β pathway activity. Luciferase report assay was performed for detection of transcriptional activity of TCF7 for Map1b; Chromatin immunoprecipitation (ChIP) was engaged for the binding of TCF7 to Map1b promoter region. RESULTS We found MACF1 enhanced MC3T3-E1 cell and C-MSCs migration in vitro through promoting microtubule (MT) stability and dynamics, and increased the injected MC3T3-E1 cell number on bone formation surface, which indicated a promoted bone formation. We further authenticated that MAP1B had a similar function to MACF1 and was regulated by MACF1 in osteogenic cell, and silencing map1b repressed MC3T3-E1 cell migration in vitro. Mechanistically, by adopting MC3T3-E1 cell with different MACF1 level or treated with LiCl/Wort, we discovered that MACF1 decreased the levels of 1265 threonine phosphorylated MAP1B (p[T1265] MAP1B) through inhibiting GSK3β activity. Additionally, total MAP1B mRNA expression level was upregulated by MACF1 through strengthening the binding of TCF7 to the map1b promoter sequence. CONCLUSION Our study uncovered a novel role of MACF1 in bone formation and MAP1B regulation, which suggested that MACF1 could be a potential therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Peihong Su
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China; Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Ye Tian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Chong Yin
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China; Department of Clinical Laboratory, Academician (expert) Workstation, Lab of Epigenetics and RNA Therapy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Xue Wang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Dijie Li
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China; Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Chaofei Yang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Jiawei Pei
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Xiaoni Deng
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Sarah King
- The University of Chicago, Ben May Department for Cancer Research, Chicago, IL 60637, USA
| | - Yu Li
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.
| | - Airong Qian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.
| |
Collapse
|
7
|
Yin C, Tian Y, Hu L, Yu Y, Wu Z, Zhang Y, Wang X, Miao Z, Qian A. MACF1 alleviates aging-related osteoporosis via HES1. J Cell Mol Med 2021; 25:6242-6257. [PMID: 34133068 PMCID: PMC8366449 DOI: 10.1111/jcmm.16579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/24/2021] [Accepted: 04/08/2021] [Indexed: 12/30/2022] Open
Abstract
Ageing-related osteoporosis is becoming an emerging threat to human health along with the ageing of human population. The decreased rate of osteogenic differentiation and bone formation is the major cause of ageing-related osteoporosis. Microtubule actin cross-linking factor 1 (MACF1) is an important cytoskeletal factor that promotes osteogenic differentiation and bone formation. However, the relationship between MACF1 expression and ageing-related osteoporosis remains unclear. This study has investigated the expression pattern of MACF1 in bone tissues of ageing-related osteoporosis patients and ageing mice. The study has further elucidated the mechanism of MACF1 promoting bone formation by inhibiting HES1 expression and activity. Moreover, the therapeutic effect of MACF1 on ageing-related osteoporosis and post-menopausal osteoporosis was evaluated through in situ injection of the MACF1 overexpression plasmid. The study supplemented the molecular mechanisms between ageing and bone formation, and provided novel targets and potential therapeutic strategy for ageing-related osteoporosis.
Collapse
Affiliation(s)
- Chong Yin
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Lab of Epigenetics and RNA TherapyDepartment of Clinical Laboratory, Academician (Expert) WorkstationAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
- Department of Laboratory MedicineNorth Sichuan Medical CollegeNanchongChina
- Translational Medicine Research CenterNorth Sichuan Medical CollegeNanchongChina
| | - Ye Tian
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Lifang Hu
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Yang Yu
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theranostics)School of PharmacyTianjin Medical UniversityTianjinChina
| | - Zixiang Wu
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Yan Zhang
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Xue Wang
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Zhiping Miao
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Airong Qian
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| |
Collapse
|
8
|
MACF1 promotes osteoblast differentiation by sequestering repressors in cytoplasm. Cell Death Differ 2021; 28:2160-2178. [PMID: 33664480 PMCID: PMC8257666 DOI: 10.1038/s41418-021-00744-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoblast differentiation leading to bone formation requires a coordinated transcriptional program. Osteoblastic cells with low level of microtubule actin crosslinking factor 1 (MACF1) show reduced osteoblast differentiation ability, however, the comprehensive mechanism of MACF1's action remains unexplored. In the current study, we found that MACF1 knockdown suppressed osteoblast differentiation by altering the transcriptome dynamics. We further identified two MACF1-interacted proteins, cyclin-dependent kinase 12 (CDK12) and MYST/Esa1-associated factor 6 (MEAF6), and two MACF1-interacted transcription factors (TFs), transcription factor 12 (TCF12) and E2F transcription factor 6 (E2F6), which repress osteoblast differentiation by altering the expression of osteogenic TFs and genes. Moreover, we found that MACF1 regulated cytoplasmic-nuclear localization of itself, TCF12 and E2F6 in a concentration-dependent manner. MACF1 oppositely regulates the expression of TCF12 and transcription factor 7 (TCF7), two TFs that drive osteoblast differentiation to opposite directions. This study reveals that MACF1, a cytoskeletal protein, acts as a sponge for repressors of osteoblast differentiation to promote osteoblast differentiation and contributes to a novel mechanistic insight of osteoblast differentiation and transcription dynamics.
Collapse
|
9
|
Cusseddu R, Robert A, Côté JF. Strength Through Unity: The Power of the Mega-Scaffold MACF1. Front Cell Dev Biol 2021; 9:641727. [PMID: 33816492 PMCID: PMC8012552 DOI: 10.3389/fcell.2021.641727] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/23/2021] [Indexed: 12/26/2022] Open
Abstract
The tight coordination of diverse cytoskeleton elements is required to support several dynamic cellular processes involved in development and tissue homeostasis. The spectraplakin-family of proteins are composed of multiple domains that provide versatility to connect different components of the cytoskeleton, including the actin microfilaments, microtubules and intermediates filaments. Spectraplakins act as orchestrators of precise cytoskeletal dynamic events. In this review, we focus on the prototypical spectraplakin MACF1, a protein scaffold of more than 700 kDa that coordinates the crosstalk between actin microfilaments and microtubules to support cell-cell connections, cell polarity, vesicular transport, proliferation, and cell migration. We will review over two decades of research aimed at understanding the molecular, physiological and pathological roles of MACF1, with a focus on its roles in developmental and cancer. A deeper understanding of MACF1 is currently limited by technical challenges associated to the study of such a large protein and we discuss ideas to advance the field.
Collapse
Affiliation(s)
- Rebecca Cusseddu
- Montreal Clinical Research Institute, Montreal, QC, Canada
- Molecular Biology Programs, Université de Montréal, Montreal, QC, Canada
| | - Amélie Robert
- Montreal Clinical Research Institute, Montreal, QC, Canada
| | - Jean-François Côté
- Montreal Clinical Research Institute, Montreal, QC, Canada
- Molecular Biology Programs, Université de Montréal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| |
Collapse
|
10
|
Gao Y, Patil S, Qian A. The Role of MicroRNAs in Bone Metabolism and Disease. Int J Mol Sci 2020; 21:ijms21176081. [PMID: 32846921 PMCID: PMC7503277 DOI: 10.3390/ijms21176081] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Bone metabolism is an intricate process involving various bone cells, signaling pathways, cytokines, hormones, growth factors, etc., and the slightest deviation can result in various bone disorders including osteoporosis, arthropathy, and avascular necrosis of femoral head. Osteoporosis is one of the most prevalent disorders affecting the skeleton, which is characterized by low bone mass and bone mineral density caused by the disruption in the balanced process of bone formation and bone resorption. The current pharmaceutical treatments such as bisphosphonates, selective estrogen receptor modulator, calcitonin, teriparatide, etc., could decrease the risk of fractures but have side-effects that have limited their long term applications. MicroRNAs (miRNAs) are one of many non-coding RNAs. These are single-stranded with a length of 19–25 nucleotides and can influence various cellular processes and play an important role in various diseases. Therefore, in this article, we review the different functions of different miRNA in bone metabolism and osteoporosis to understand their mechanism of action for the development of possible therapeutics.
Collapse
Affiliation(s)
- Yongguang Gao
- Laboratory for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (Y.G.); (S.P.)
- Department of Chemistry, Tangshan Normal University, Tangshan 063000, China
| | - Suryaji Patil
- Laboratory for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (Y.G.); (S.P.)
| | - Airong Qian
- Laboratory for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (Y.G.); (S.P.)
- Correspondence: ; Tel.: +86-135-7210-8260
| |
Collapse
|
11
|
Su P, Yin C, Li D, Yang C, Wang X, Pei J, Tian Y, Qian A. MACF1 promotes preosteoblast migration by mediating focal adhesion turnover through EB1. Biol Open 2020; 9:bio048173. [PMID: 32139394 PMCID: PMC7104863 DOI: 10.1242/bio.048173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/20/2020] [Indexed: 12/25/2022] Open
Abstract
Microtubule actin crosslinking factor 1 (MACF1) is a widely expressed cytoskeletal linker and plays an essential role in various cells' functions by mediating cytoskeleton organization and dynamics. However, the role of MACF1 on preosteoblast migration is not clear. Here, by using MACF1 knockdown and overexpressed MC3T3-E1 cells, we found MACF1 positively regulated preosteoblast migration induced by cell polarization. Furthermore, immunofluorescent staining showed that MACF1 increased end-binding protein (EB1) distribution on microtubule (MT), and decreased EB1 distribution on focal adhesion (FA) complex. Moreover, upregulation of MACF1 activated Src level and enhanced the colocalization of EB1 with activated Src. In addition, MACF1 diminished colocalization of EB1 with adenomatous polyposis coli (APC), which induced EB1 release from FA and promoted FA turnover. These results indicated an important role and mechanism of MACF1 in regulating preosteoblast migration through promoting FA turnover by mediating EB1 colocalization with Src and APC, which inferred that MACF1 might be a potential target for preventing and treating bone disorders.
Collapse
Affiliation(s)
- Peihong Su
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Chong Yin
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Dijie Li
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Chaofei Yang
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Xue Wang
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Jiawei Pei
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Ye Tian
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Airong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
12
|
Mesenchymal MACF1 Facilitates SMAD7 Nuclear Translocation to Drive Bone Formation. Cells 2020; 9:cells9030616. [PMID: 32143362 PMCID: PMC7140458 DOI: 10.3390/cells9030616] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/27/2020] [Accepted: 02/29/2020] [Indexed: 02/07/2023] Open
Abstract
Microtubule actin crosslinking factor 1 (MACF1) is a large crosslinker that contributes to cell integrity and cell differentiation. Recent studies show that MACF1 is involved in multiple cellular functions such as neuron development and epidermal migration, and is the molecular basis for many degenerative diseases. MACF1 is highly abundant in bones, especially in mesenchymal stem cells; however, its regulatory role is still less understood in bone formation and degenerative bone diseases. In this study, we found MACF1 expression in mesenchymal stem cells (MSCs) of osteoporotic bone specimens was significantly lower. By conditional gene targeting to delete the mesenchymal Macf1 gene in mice, we observed in MSCs decreased osteogenic differentiation capability. During early stage bone development, the MACF1 conditional knockout (cKO) mice exhibit significant ossification retardation in skull and hindlimb, and by adulthood, mesenchymal loss of MACF1 attenuated bone mass, bone microarchitecture, and bone formation capability significantly. Further, we showed that MACF1 interacts directly with SMAD family member 7 (SMAD7) and facilitates SMAD7 nuclear translocation to initiate downstream osteogenic pathways. Hopefully these findings will expand the biological scope of the MACF1 gene, and provide an experimental basis for targeting MACF1 in degenerative bone diseases such as osteoporosis.
Collapse
|
13
|
Bonner K, Borlay D, Kutten O, Quick QA. Inhibition of the Spectraplakin Protein Microtubule Actin Crosslinking Factor 1 Sensitizes Glioblastomas to Radiation. Brain Tumor Res Treat 2020; 8:43-52. [PMID: 32390353 PMCID: PMC7221465 DOI: 10.14791/btrt.2020.8.e1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 10/29/2019] [Accepted: 02/24/2020] [Indexed: 12/22/2022] Open
Abstract
Background Microtubule actin crosslinking factor 1 (MACF1) is a spectraplakin cytoskeletal crosslinking protein whose function and role in cancer biology has lacked investigation. Recent studies have identified MACF1 as a novel target in glioblastomas expressed in tissue from tumor patient explants but not normal brain tissue and when silenced has an antitumorigenic impact on these tumors. Radiation as a single agent therapy to treat glioblastomas has been used for decades and has done little to improve survival of individuals diagnosed with this disease. However, contemporary clinical radiotherapy protocols have provided evidence that combinatorial radiotherapy approaches confer a therapeutic benefit in glioblastoma patients. In this study MACF1 was investigated as a radiosensitization target in glioblastomas. Methods To provide context of MACF1 in glioblastomas, The Cancer Genome Atlas expression analyses were performed in conjunction with genes associated with glioblastoma evolution, while a genetic inhibitory approach, cell migratory assays, and immunofluorescence procedures were used to evaluate responses to MACF1 suppression with radiation. Additionally, expression analyses were conducted to assess co-expression of mTOR signaling pathway regulators and MACF1 in glioblastoma patient samples. Results Our amalgamation approach demonstrated that negative regulation of MACF1, which was positively correlated with epidermal growth factor receptor and p70s6k expression, enhanced the sensitivity of glioblastoma cells to radiation as a consequence of reducing glioblastoma cell viability and migration. Mechanistically, the antitumorigenic effects on glioblastoma cell behaviors after radiation and impairing MACF1 function were associated with decreased expression of ribosomal protein S6, a downstream effector of p70s6k. Conclusion MACF1 represents a diagnostic marker with target specificity in glioblastomas that can enhance the efficacy of radiation while minimizing normal tissue toxicity. This approach could potentially expand combinatorial radiation strategies for glioblastoma treatments via impairment of translational regulatory processes that contribute to poor patient survival.
Collapse
Affiliation(s)
- Kala Bonner
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | - Danielle Borlay
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | - Orica Kutten
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | - Quincy A Quick
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA.
| |
Collapse
|
14
|
Li D, Tian Y, Yin C, Huai Y, Zhao Y, Su P, Wang X, Pei J, Zhang K, Yang C, Dang K, Jiang S, Miao Z, Li M, Hao Q, Zhang G, Qian A. Silencing of lncRNA AK045490 Promotes Osteoblast Differentiation and Bone Formation via β-Catenin/TCF1/Runx2 Signaling Axis. Int J Mol Sci 2019; 20:ijms20246229. [PMID: 31835596 PMCID: PMC6941011 DOI: 10.3390/ijms20246229] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis, a disease characterized by both loss of bone mass and structural deterioration of bone, is the most common reason for a broken bone among the elderly. It is known that the attenuated differentiation ability of osteogenic cells has been regarded as one of the greatest contributors to age-related bone formation reduction. However, the effects of current therapies are still unsatisfactory. In this study we identify a novel long noncoding RNA AK045490 which is correlated with osteogenic differentiation and enriched in skeletal tissues of mice. In vitro analysis of bone-derived mesenchymal stem cells (BMSCs) showed that AK045490 inhibited osteoblast differentiation. In vivo inhibition of AK045490 by its small interfering RNA rescued bone formation in ovariectomized osteoporosis mice model. Mechanistically, AK045490 inhibited the nuclear translocation of β-catenin and downregulated the expression of TCF1, LEF1, and Runx2. The results suggest that Lnc-AK045490 suppresses β-catenin/TCF1/Runx2 signaling and inhibits osteoblast differentiation and bone formation, providing a novel mechanism of osteogenic differentiation and a potential drug target for osteoporosis.
Collapse
Affiliation(s)
- Dijie Li
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, SAR, China
| | - Ye Tian
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Chong Yin
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Ying Huai
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Yipu Zhao
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Peihong Su
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Xue Wang
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Jiawei Pei
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Kewen Zhang
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Chaofei Yang
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Kai Dang
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Shanfeng Jiang
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Zhiping Miao
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi’an 710032, China; (M.L.); (Q.H.)
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi’an 710032, China; (M.L.); (Q.H.)
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, SAR, China
- Correspondence: (G.Z.); (A.Q.); Tel.: +86-29-88491840 (G.Z. & A.Q.)
| | - Airong Qian
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (D.L.); (Y.T.); (C.Y.); (Y.H.); (Y.Z.); (P.S.); (X.W.); (J.P.); (K.Z.); (C.Y.); (K.D.); (Z.M.)
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Correspondence: (G.Z.); (A.Q.); Tel.: +86-29-88491840 (G.Z. & A.Q.)
| |
Collapse
|
15
|
Qiu WX, Ma XL, Lin X, Zhao F, Li DJ, Chen ZH, Zhang KW, Zhang R, Wang P, Xiao YY, Miao ZP, Dang K, Wu XY, Qian AR. Deficiency of Macf1 in osterix expressing cells decreases bone formation by Bmp2/Smad/Runx2 pathway. J Cell Mol Med 2019; 24:317-327. [PMID: 31709715 PMCID: PMC6933318 DOI: 10.1111/jcmm.14729] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/16/2019] [Accepted: 10/03/2019] [Indexed: 12/18/2022] Open
Abstract
Microtubule actin cross-linking factor 1 (Macf1) is a spectraplakin family member known to regulate cytoskeletal dynamics, cell migration, neuronal growth and cell signal transduction. We previously demonstrated that knockdown of Macf1 inhibited the differentiation of MC3T3-E1 cell line. However, whether Macf1 could regulate bone formation in vivo is unclear. To study the function and mechanism of Macf1 in bone formation and osteogenic differentiation, we established osteoblast-specific Osterix (Osx) promoter-driven Macf1 conditional knockout mice (Macf1f/f Osx-Cre). The Macf1f/f Osx-Cre mice displayed delayed ossification and decreased bone mass. Morphological and mechanical studies showed deteriorated trabecular microarchitecture and impaired biomechanical strength of femur in Macf1f/f Osx-Cre mice. In addition, the differentiation of primary osteoblasts isolated from calvaria was inhibited in Macf1f/f Osx-Cre mice. Deficiency of Macf1 in primary osteoblasts inhibited the expression of osteogenic marker genes (Col1, Runx2 and Alp) and the number of mineralized nodules. Furthermore, deficiency of Macf1 attenuated Bmp2/Smad/Runx2 signalling in primary osteoblasts of Macf1f/f Osx-Cre mice. Together, these results indicated that Macf1 plays a significant role in bone formation and osteoblast differentiation by regulating Bmp2/Smad/Runx2 pathway, suggesting that Macf1 might be a therapeutic target for bone disease.
Collapse
Affiliation(s)
- Wu-Xia Qiu
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xiao-Li Ma
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xiao Lin
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Fan Zhao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Di-Jie Li
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Zhi-Hao Chen
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Ke-Wen Zhang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Ru Zhang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Pai Wang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yun-Yun Xiao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Zhi-Ping Miao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Kai Dang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xiao-Yang Wu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Ai-Rong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
16
|
Zhang Y, Yin C, Hu L, Chen Z, Zhao F, Li D, Ma J, Ma X, Su P, Qiu W, Yang C, Wang P, Li S, Zhang G, Wang L, Qian A, Xian CJ. MACF1 Overexpression by Transfecting the 21 kbp Large Plasmid PEGFP-C1A-ACF7 Promotes Osteoblast Differentiation and Bone Formation. Hum Gene Ther 2019; 29:259-270. [PMID: 29334773 DOI: 10.1089/hum.2017.153] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Microtubule actin crosslinking factor 1 (MACF1) is a large spectraplakin protein known to have crucial roles in regulating cytoskeletal dynamics, cell migration, growth, and differentiation. However, its role and action mechanism in bone remain unclear. The present study investigated optimal conditions for effective transfection of the large plasmid PEGFP-C1A-ACF7 (∼21 kbp) containing full-length human MACF1 cDNA, as well as the potential role of MACF1 in bone formation. To enhance MACF1 expression, the plasmid was transfected into osteogenic cells by electroporation in vitro and into mouse calvaria with nanoparticles. Then, transfection efficiency, osteogenic marker expression, calvarial thickness, and bone formation were analyzed. Notably, MACF1 overexpression triggered a drastic increase in osteogenic gene expression, alkaline phosphatase activity, and matrix mineralization in vitro. Mouse calvarial thickness, mineral apposition rate, and osteogenic marker protein expression were significantly enhanced by local transfection. In addition, MACF1 overexpression promoted β-catenin expression and signaling. In conclusion, MACF1 overexpression by transfecting the large plasmid containing full-length MACF1 cDNA promotes osteoblast differentiation and bone formation via β-catenin signaling. Current data will provide useful experimental parameters for the transfection of large plasmids and a novel strategy based on promoting bone formation for prevention and therapy of bone disorders.
Collapse
Affiliation(s)
- Yan Zhang
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Chong Yin
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Lifang Hu
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Zhihao Chen
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Fan Zhao
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Dijie Li
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Jianhua Ma
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Xiaoli Ma
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Peihong Su
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Wuxia Qiu
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Chaofei Yang
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Pai Wang
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Siyu Li
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Ge Zhang
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Liping Wang
- 4 Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia , Adelaide, Australia
| | - Airong Qian
- 1 Bone Metabolism Lab, School of Life Sciences, Northwestern Polytechnical University , Xi'an, Shaanxi, China
- 2 Shenzhen Research Institute of Northwestern Polytechnical University , Shenzhen, Guangdong, China
- 3 NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University , Xi'an, Shaanxi, China
| | - Cory J Xian
- 4 Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia , Adelaide, Australia
| |
Collapse
|
17
|
Yin C, Tian Y, Yu Y, Wang H, Wu Z, Huang Z, Zhang Y, Li D, Yang C, Wang X, Li Y, Qian A. A novel long noncoding RNA AK016739 inhibits osteoblast differentiation and bone formation. J Cell Physiol 2019; 234:11524-11536. [PMID: 30656695 DOI: 10.1002/jcp.27815] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 11/01/2018] [Indexed: 01/04/2023]
Abstract
The incidence of postmenopausal osteoporosis research 50% in middle-aged and older women, however, effects of existing therapy are not ideal. Emerging evidence have proved that long noncoding RNAs (lncRNAs) was correlated with multiple physiological and pathology processes including development, carcinogenesis, and osteogenesis. However, reports on lncRNAs regulating bone formation were relatively limited. In this study, we screened osteogenic lncRNAs through mRNA/lncRNA microarray combined with gene coexpression analysis. The biological function of the screened lncRNA was assessed both in vitro and in vivo. The effects of the lncRNA on osteogenic transcription factors were also evaluated. We identified AK016739, which was correlated with osteogenic differentiation and enriched in skeletal tissues of mice. The expression levels of AK016739 in bone-derived mesenchymal stem cells were increased with age and negatively correlated with osteogenic differentiation marker genes. Experiments showed that AK016739 inhibited osteoblast differentiation, and in vivo inhibition of AK016739 by its small interfering RNA would rescue bone formation in ovariectomized osteoporosis mice model. In addition, AK016739 suppressed both expression levels and activities of osteogenic transcription factors. This newly identified lncRNA AK016739 has revealed a new mechanism of osteogenic differentiation and provided new targets for treatment of skeletal disorders.
Collapse
Affiliation(s)
- Chong Yin
- Laboratory for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Ye Tian
- Laboratory for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yang Yu
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Haoyu Wang
- Department of Software Technology and Service Engineering, School of Software and Microelectronics, Peking University, Beijing, China
| | - Zhixiang Wu
- Laboratory for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Zizhan Huang
- Laboratory for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yan Zhang
- Laboratory for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Dijie Li
- Laboratory for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Chaofei Yang
- Laboratory for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xue Wang
- Laboratory for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yu Li
- Laboratory for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Airong Qian
- Laboratory for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
18
|
PEP-1-PEA15 suppresses inflammatory responses by regulation of MAPK in macrophages and animal models. Immunobiology 2018; 223:709-717. [DOI: 10.1016/j.imbio.2018.07.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/10/2018] [Accepted: 07/17/2018] [Indexed: 12/19/2022]
|
19
|
Hu L, Huang Z, Wu Z, Ali A, Qian A. Mammalian Plakins, Giant Cytolinkers: Versatile Biological Functions and Roles in Cancer. Int J Mol Sci 2018; 19:ijms19040974. [PMID: 29587367 PMCID: PMC5979291 DOI: 10.3390/ijms19040974] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/18/2018] [Accepted: 03/20/2018] [Indexed: 01/07/2023] Open
Abstract
Cancer is a highly lethal disease that is characterized by aberrant cell proliferation, migration, and adhesion, which are closely related to the dynamic changes of cytoskeletons and cytoskeletal-adhesion. These will further result in cell invasion and metastasis. Plakins are a family of giant cytolinkers that connect cytoskeletal elements with each other and to junctional complexes. With various isoforms composed of different domain structures, mammalian plakins are broadly expressed in numerous tissues. They play critical roles in many cellular processes, including cell proliferation, migration, adhesion, and signaling transduction. As these cellular processes are key steps in cancer development, mammalian plakins have in recent years attracted more and more attention for their potential roles in cancer. Current evidence shows the importance of mammalian plakins in various human cancers and demonstrates mammalian plakins as potential biomarkers for cancer. Here, we introduce the basic characteristics of mammalian plakins, review the recent advances in understanding their biological functions, and highlight their roles in human cancers, based on studies performed by us and others. This will provide researchers with a comprehensive understanding of mammalian plakins, new insights into the development of cancer, and novel targets for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Lifang Hu
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Zizhan Huang
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Zixiang Wu
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Arshad Ali
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Airong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| |
Collapse
|
20
|
Yin C, Zhang Y, Hu L, Tian Y, Chen Z, Li D, Zhao F, Su P, Ma X, Zhang G, Miao Z, Wang L, Qian A, Xian CJ. Mechanical unloading reduces microtubule actin crosslinking factor 1 expression to inhibit β-catenin signaling and osteoblast proliferation. J Cell Physiol 2018; 233:5405-5419. [PMID: 29219183 DOI: 10.1002/jcp.26374] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/29/2017] [Indexed: 02/03/2023]
Abstract
Mechanical unloading was considered a major threat to bone homeostasis, and has been shown to decrease osteoblast proliferation although the underlying mechanism is unclear. Microtubule actin crosslinking factor 1 (MACF1) is a cytoskeletal protein that regulates cellular processes and Wnt/β-catenin pathway, an essential signaling pathway for osteoblasts. However, the relationship between MACF1 expression and mechanical unloading, and the function and the associated mechanisms of MACF1 in regulating osteoblast proliferation are unclear. This study investigated effects of mechanical unloading on MACF1 expression levels in cultured MC3T3-E1 osteoblastic cells and in femurs of mice with hind limb unloading; and it also examined the role and potential action mechanisms of MACF1 in osteoblast proliferation in MACF1-knockdown, overexpressed or control MC3T3-E1 cells treated with or without the mechanical unloading condition. Results showed that the mechanical unloading condition inhibited osteoblast proliferation and MACF1 expression in MC3T3-E1 osteoblastic cells and mouse femurs. MACF1 knockdown decreased osteoblast proliferation, while MACF1 overexpression increased it. The inhibitory effect of mechanical unloading on osteoblast proliferation also changed with MACF1 expression levels. Furthermore, MACF1 was found to enhance β-catenin expression and activity, and mechanical unloading decreased β-catenin expression through MACF1. Moreover, β-catenin was found an important regulator of osteoblast proliferation, as its preservation by treatment with its agonist lithium attenuated the inhibitory effects of MACF1-knockdown or mechanical unloading on osteoblast proliferation. Taken together, mechanical unloading decreases MACF1 expression, and MACF1 up-regulates osteoblast proliferation through enhancing β-catenin signaling. This study has thus provided a mechanism for mechanical unloading-induced inhibited osteoblast proliferation.
Collapse
Affiliation(s)
- Chong Yin
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Yan Zhang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Lifang Hu
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Ye Tian
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Zhihao Chen
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Dijie Li
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Fan Zhao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Peihong Su
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Xiaoli Ma
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China
| | - Ge Zhang
- Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Zhiping Miao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Liping Wang
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Airong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
21
|
Miao Z, Ali A, Hu L, Zhao F, Yin C, Chen C, Yang T, Qian A. Microtubule actin cross-linking factor 1, a novel potential target in cancer. Cancer Sci 2017; 108:1953-1958. [PMID: 28782898 PMCID: PMC5623738 DOI: 10.1111/cas.13344] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/28/2017] [Accepted: 08/02/2017] [Indexed: 01/09/2023] Open
Abstract
Cancer is a polygenic disease characterized by uncontrolled growth of normal body cells, deregulation of the cell cycle as well as resistance to apoptosis. The spectraplakin protein microtubule actin cross-linking factor 1 (MACF1) plays an essential function in various cellular processes, including cell proliferation, migration, signaling transduction and embryo development. MACF1 is also involved in processes such as metastatic invasion in which cytoskeleton organization is a critical element that contributes to tumor progression in various human cancers. Aberrant expression of MACF1 initiates the tumor cell proliferation, and migration and metastasis in numerous cancers, such as breast cancer, colon cancer, lung cancer and glioblastoma. In this review, we summarized the current knowledge of MACF1 and its critical role in different human cancers. This will be helpful for researchers to investigate the novel functional role of MACF1 in human cancers and as a potential target to enhance the efficacy of therapeutic treatment modalities.
Collapse
Affiliation(s)
- Zhiping Miao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institution of Northwestern Polytechnical University, Shenzhen, China
| | - Arshad Ali
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institution of Northwestern Polytechnical University, Shenzhen, China
| | - Lifang Hu
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institution of Northwestern Polytechnical University, Shenzhen, China
| | - Fan Zhao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institution of Northwestern Polytechnical University, Shenzhen, China
| | - Chong Yin
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institution of Northwestern Polytechnical University, Shenzhen, China
| | - Chu Chen
- Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Tuanmin Yang
- Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Airong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institution of Northwestern Polytechnical University, Shenzhen, China
| |
Collapse
|
22
|
Hu L, Su P, Yin C, Zhang Y, Li R, Yan K, Chen Z, Li D, Zhang G, Wang L, Miao Z, Qian A, Xian CJ. Microtubule actin crosslinking factor 1 promotes osteoblast differentiation by promoting β-catenin/TCF1/Runx2 signaling axis. J Cell Physiol 2017. [PMID: 28621459 DOI: 10.1002/jcp.26059] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Osteoblast differentiation is a multistep process delicately regulated by many factors, including cytoskeletal dynamics and signaling pathways. Microtubule actin crosslinking factor 1 (MACF1), a key cytoskeletal linker, has been shown to play key roles in signal transduction and in diverse cellular processes; however, its role in regulating osteoblast differentiation is still needed to be elucidated. To further uncover the functions and mechanisms of action of MACF1 in osteoblast differentiation, we examined effects of MACF1 knockdown (MACF1-KD) in MC3T3-E1 osteoblastic cells on their osteoblast differentiation and associated molecular mechanisms. The results showed that knockdown of MACF1 significantly suppressed mineralization of MC3T3-E1 cells, down-regulated the expression of key osteogenic genes alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2) and type I collagen α1 (Col Iα1). Knockdown of MACF1 dramatically reduced the nuclear translocation of β-catenin, decreased the transcriptional activation of T cell factor 1 (TCF1), and down-regulated the expression of TCF1, lymphoid enhancer-binding factor 1 (LEF1), and Runx2, a target gene of β-catenin/TCF1. In addition, MACF1-KD increased the active level of glycogen synthase kinase-3β (GSK-3β), which is a key regulator for β-catenin signal transduction. Moreover, the reduction of nuclear β-catenin amount and decreased expression of TCF1 and Runx2 were significantly reversed in MACF1-KD cells when treated with lithium chloride, an agonist for β-catenin by inhibiting GSK-3β activity. Taken together, these findings suggest that knockdown of MACF1 in osteoblastic cells inhibits osteoblast differentiation through suppressing the β-catenin/TCF1-Runx2 axis. Thus, a novel role of MACF1 in and a new mechanistic insight of osteoblast differentiation are uncovered.
Collapse
Affiliation(s)
- Lifang Hu
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Peihong Su
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Chong Yin
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Yan Zhang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Runzhi Li
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Kun Yan
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Zhihao Chen
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Dijie Li
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Ge Zhang
- Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Liping Wang
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Zhiping Miao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Airong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.,Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, China.,NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an, China
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
23
|
Moffat JJ, Ka M, Jung EM, Smith AL, Kim WY. The role of MACF1 in nervous system development and maintenance. Semin Cell Dev Biol 2017; 69:9-17. [PMID: 28579452 DOI: 10.1016/j.semcdb.2017.05.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/12/2017] [Accepted: 05/29/2017] [Indexed: 12/14/2022]
Abstract
Microtubule-actin crosslinking factor 1 (MACF1), also known as actin crosslinking factor 7 (ACF7), is essential for proper modulation of actin and microtubule cytoskeletal networks. Most MACF1 isoforms are expressed broadly in the body, but some are exclusively found in the nervous system. Consequentially, MACF1 is integrally involved in multiple neural processes during development and in adulthood, including neurite outgrowth and neuronal migration. Furthermore, MACF1 participates in several signaling pathways, including the Wnt/β-catenin and GSK-3 signaling pathways, which regulate key cellular processes, such as proliferation and cell migration. Genetic mutation or dysregulation of the MACF1 gene has been associated with neurodevelopmental and neurodegenerative diseases, specifically schizophrenia and Parkinson's disease. MACF1 may also play a part in neuromuscular disorders and have a neuroprotective role in the optic nerve. In this review, the authors seek to synthesize recent findings relating to the roles of MACF1 within the nervous system and explore potential novel functions of MACF1 not yet examined.
Collapse
Affiliation(s)
- Jeffrey J Moffat
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Minhan Ka
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Eui-Man Jung
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Amanda L Smith
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Woo-Yang Kim
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
24
|
Hu L, Xiao Y, Xiong Z, Zhao F, Yin C, Zhang Y, Su P, Li D, Chen Z, Ma X, Zhang G, Qian A. MACF1, versatility in tissue-specific function and in human disease. Semin Cell Dev Biol 2017; 69:3-8. [PMID: 28577926 DOI: 10.1016/j.semcdb.2017.05.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/18/2017] [Accepted: 05/26/2017] [Indexed: 01/24/2023]
Abstract
Spectraplakins are a family of evolutionarily conserved gigantic proteins and play critical roles in many cytoskeleton-related processes. Microtubule actin crosslinking factor 1 (MACF1) is one of the most versatile spectraplakin with multiple isoforms. As a broadly expressed mammalian spectraplakin, MACF1 is important in maintaining normal functions of many tissues. The loss-of-function studies using knockout mouse models reveal the pivotal roles of MACF1 in embryo development, skin integrity maintenance, neural development, bone formation, and colonic paracellular permeability. Mutation in the human MACF1 gene causes a novel myopathy genetic disease. In addition, abnormal expression of MACF1 is associated with schizophrenia, Parkinson's disease, cancer and osteoporosis. This demonstrates the crucial roles of MACF1 in physiology and pathology. Here, we review the research advances of MACF1's roles in specific tissue and in human diseases, providing the perspectives of MACF1 for future studies.
Collapse
Affiliation(s)
- Lifang Hu
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, 518057, China; NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yunyun Xiao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, 518057, China; NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China
| | - Zhipeng Xiong
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, 518057, China; NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China
| | - Fan Zhao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, 518057, China; NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China
| | - Chong Yin
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, 518057, China; NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yan Zhang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, 518057, China; NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China
| | - Peihong Su
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, 518057, China; NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China
| | - Dijie Li
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, 518057, China; NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China
| | - Zhihao Chen
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, 518057, China; NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China
| | - Xiaoli Ma
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, 518057, China; NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China
| | - Ge Zhang
- NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China; Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Airong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, 518057, China; NPU-HKBU Joint Research Centre for Translational Medicine on Musculoskeletal Health in Space, Northwestern Polytechnical University, Xi'an 710072, China.
| |
Collapse
|
25
|
Son MJ, Kim WK, Oh KJ, Park A, Lee DS, Han BS, Lee SC, Bae KH. Methyltransferase and demethylase profiling studies during brown adipocyte differentiation. BMB Rep 2017; 49:388-93. [PMID: 27157542 PMCID: PMC5032007 DOI: 10.5483/bmbrep.2016.49.7.062] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Indexed: 02/03/2023] Open
Abstract
Although brown adipose tissue is important with regard to energy balance, the molecular mechanism of brown adipocyte differentiation has not been extensively studied. Specifically, regulation factors at the level of protein modification are largely unknown. In this study, we examine the changes in the expression level of enzymes which are involved in protein lysine methylation during brown adipocyte differentiation. Several enzymes, in this case SUV420H2, PRDM9, MLL3 and JHDM1D, were found to be up-regulated. On the other hand, Set7/9 was significantly down-regulated. In the case of SUV420H2, the expression level increased sharply during brown adipocyte differentiation, whereas the expression of SUV420H2 was marginally enhanced during the white adipocyte differentiation. The knock-down of SUV420H2 caused the suppression of brown adipocyte differentiation, as compared to a scrambled control. These results suggest that SUV420H2, a methyltransferase, is involved in brown adipocyte differentiation, and that the methylation of protein lysine is important in brown adipocyte differentiation. [BMB Reports 2016; 49(7): 388-393]
Collapse
Affiliation(s)
- Min Jeong Son
- Metabolic Regulation Research Center, KRIBB, Daejeon 34141, Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, KRIBB; Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, KRIBB, Daejeon 34141, Korea
| | - Anna Park
- Metabolic Regulation Research Center, KRIBB, Daejeon 34141, Korea
| | - Da Som Lee
- Metabolic Regulation Research Center, KRIBB, Daejeon 34141, Korea
| | - Baek Soo Han
- Metabolic Regulation Research Center, KRIBB; Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, KRIBB; Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, KRIBB; Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea
| |
Collapse
|
26
|
Kim N, Min WK, Park MH, Lee JK, Jin HK, Bae JS. Neuropeptide Y protects kidney against cisplatin-induced nephrotoxicity by regulating p53-dependent apoptosis pathway. BMB Rep 2017; 49:288-92. [PMID: 26728272 PMCID: PMC5070709 DOI: 10.5483/bmbrep.2016.49.5.231] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Indexed: 11/23/2022] Open
Abstract
Cisplatin is a platinum-based chemotherapeutic drug for treating various types of cancers. However, the use of cisplatin is limited by its negative effect on normal tissues, particularly nephrotoxicity. Various mechanisms such as DNA adduct formation, mitochondrial dysfunction, oxidative stress, and apoptosis are involved in the adverse effect induced by cisplatin treatment. Several studies have suggested that neuropeptide Y (NPY) is involved in neuroprotection as well as restoration of bone marrow dysfunction from chemotherapy induced nerve injury. However, the role of NPY in chemotherapy-induced nephrotoxicity has not been studied. Here, we show that NPY rescues renal dysfunction by reducing the expression of pro-apoptotic proteins in cisplatin induced nephrotoxicity through Y1 receptor, suggesting that NPY can protect kidney against cisplatin nephrotoxicity as a possible useful agent to prevent and treat cisplatin-induced nephrotoxicity. [BMB Reports 2016; 49(5): 288-292]
Collapse
Affiliation(s)
- Namoh Kim
- Stem Cell Neuroplasticity Research Group, Kyungpook National University; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| | - Woo-Kie Min
- Department of Orthopaedic Surgery, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Min Hee Park
- Stem Cell Neuroplasticity Research Group, Kyungpook National University; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| | - Jong Kil Lee
- Stem Cell Neuroplasticity Research Group, Kyungpook National University; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| | - Hee Kyung Jin
- Stem Cell Neuroplasticity Research Group, Kyungpook National University; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| | - Jae-Sung Bae
- Stem Cell Neuroplasticity Research Group, Kyungpook National University; Department of Physiology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University; Department of Biomedical Science, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
27
|
Song I, Jeong BC, Choi YJ, Chung YS, Kim N. GATA4 negatively regulates bone sialoprotein expression in osteoblasts. BMB Rep 2017; 49:343-8. [PMID: 26973342 PMCID: PMC5070723 DOI: 10.5483/bmbrep.2016.49.6.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Indexed: 11/20/2022] Open
Abstract
GATA4 has been reported to act as a negative regulator in osteoblast differentiation by inhibiting the Dlx5 transactivation of Runx2 via the attenuation of the binding ability of Dlx5 to the Runx2 promoter region. Here, we determine the role of GATA4 in the regulation of bone sialoprotein (Bsp) in osteoblasts. We observed that the overexpression of Runx2 or Sox9 induced the Bsp expression in osteoblastic cells. Silencing GATA4 further enhanced the Runx2- and Sox9-mediated Bsp promoter activity, whereas GATA4 overexpression down-regulated Bsp promoter activity mediated by Runx2 and Sox9. GATA4 also interacted with Runx2 and Sox9, by attenuating the binding ability of Runx2 and Sox9 to the Bsp promoter region. Our data suggest that GATA4 acts as a negative regulator of Bsp expression in osteoblasts. [BMB Reports 2016; 49(6): 343-348]
Collapse
Affiliation(s)
- Insun Song
- Shool of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Byung-Chul Jeong
- Departments of Pharmacology and Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea
| | - Yong Jun Choi
- Department of Endocrinology, Ajou University, Suwon 16499, Korea
| | - Yoon-Sok Chung
- Department of Endocrinology, Ajou University, Suwon 16499, Korea
| | - Nacksung Kim
- Departments of Pharmacology and Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea
| |
Collapse
|
28
|
Kim E, Kim S, Lee JH, Kwon YT, Lee MJ. Ablation of Arg-tRNA-protein transferases results in defective neural tube development. BMB Rep 2017; 49:443-8. [PMID: 27345715 PMCID: PMC5070732 DOI: 10.5483/bmbrep.2016.49.8.087] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Indexed: 12/05/2022] Open
Abstract
The arginylation branch of the N-end rule pathway is a ubiquitin-mediated
proteolytic system in which post-translational conjugation of Arg by
ATE1-encoded Arg-tRNA-protein transferase to N-terminal
Asp, Glu, or oxidized Cys residues generates essential degradation signals.
Here, we characterized the ATE1−/− mice
and identified the essential role of N-terminal arginylation in neural tube
development. ATE1-null mice showed severe intracerebral
hemorrhages and cystic space near the neural tubes. Expression of ATE1 was
prominent in the developing brain and spinal cord, and this pattern overlapped
with the migration path of neural stem cells. The
ATE1−/− brain showed defective
G-protein signaling. Finally, we observed reduced mitosis in
ATE1−/− neuroepithelium and a
significantly higher nitric oxide concentration in the
ATE1−/− brain. Our results strongly
suggest that the crucial role of ATE1 in neural tube development is directly
related to proper turn-over of the RGS4 protein, which participate in the
oxygen-sensing mechanism in the cells. [BMB Reports 2016; 49(8): 443-448]
Collapse
Affiliation(s)
- Eunkyoung Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Seonmu Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jung Hoon Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Yong Tae Kwon
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Min Jae Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine; Department of Biomedical Sciences, Seoul National University Graduate School; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
29
|
Lee H, Park J, Kim EE, Yoo YS, Song EJ. Proteasome inhibitors attenuated cholesterol-induced cardiac hypertrophy in H9c2 cells. BMB Rep 2017; 49:270-5. [PMID: 26592933 PMCID: PMC5070706 DOI: 10.5483/bmbrep.2016.49.5.187] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Indexed: 11/20/2022] Open
Abstract
The Ubiquitin proteasome system (UPS) plays roles in protein degradation, cell
cycle control, and growth and inflammatory cell signaling. Dysfunction of UPS in
cardiac diseases has been seen in many studies. Cholesterol acts as an inducer
of cardiac hypertrophy. In this study, the effect of proteasome inhibitors on
the cholesterol-induced hypertrophic growth in H9c2 cells is examined in order
to observe whether UPS is involved in cardiac hypertrophy. The treatment of
proteasome inhibitors MG132 and Bortezomib markedly reduced cellular surface
area and mRNA expression of β-MHC in cholesterol-induced cardiac
hypertrophy. In addition, activated AKT and ERK were significantly attenuated by
MG132 and Bortezomib in cholesterol-induced cardiac hypertrophy. We demonstrated
that cholesterol-induced cardiac hypertrophy was suppressed by proteasome
inhibitors. Thus, regulatory mechanism of cholesterol-induced cardiac
hypertrophy by proteasome inhibitors may provide a new therapeutic strategy to
prevent the progression of heart failure. [BMB Reports 2016; 49(5): 270-275]
Collapse
Affiliation(s)
- Hyunjung Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Jinyoung Park
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Eunice EunKyeong Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Young Sook Yoo
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Eun Joo Song
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea
| |
Collapse
|
30
|
Afghani N, Mehta T, Wang J, Tang N, Skalli O, Quick QA. Microtubule actin cross-linking factor 1, a novel target in glioblastoma. Int J Oncol 2016; 50:310-316. [PMID: 27959385 DOI: 10.3892/ijo.2016.3798] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/05/2016] [Indexed: 12/26/2022] Open
Abstract
Genetic heterogeneity is recognized as a major contributing factor of glioblastoma resistance to clinical treatment modalities and consequently low overall survival rates. This genetic diversity results in variations in protein expression, both intratumorally and between individual glioblastoma patients. In this regard, the spectraplakin protein, microtubule actin cross-linking factor 1 (MACF1), was examined in glioblastoma. An expression analysis of MACF1 in various types of brain tumor tissue revealed that MACF1 was predominately present in grade III-IV astroctyomas and grade IV glioblastoma, but not in normal brain tissue, normal human astrocytes and lower grade brain tumors. Subsequent genetic inhibition experiments showed that suppression of MACF1 selectively inhibited glioblastoma cell proliferation and migration in cell lines established from patient derived xenograft mouse models and immortalized glioblastoma cell lines that were associated with downregulation of the Wnt-signaling mediators, Axin1 and β-catenin. Additionally, concomitant MACF1 silencing with the chemotherapeutic agent temozolomide (TMZ) used for the clinical treatment of glioblastomas cooperatively reduced the proliferative capacity of glioblastoma cells. In conclusion, the present study represents the first investigation on the functional role of MACF1 in tumor cell biology, as well as demonstrates its potential as a unique biomarker that can be targeted synergistically with TMZ as part of a combinatorial therapeutic approach for the treatment of genetically multifarious glioblastomas.
Collapse
Affiliation(s)
- Najlaa Afghani
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | - Toral Mehta
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| | - Jialiang Wang
- Department of Neurological Surgery, Vanderbilt Medical Center, Nashville, TN, USA
| | - Nan Tang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Omar Skalli
- Department of Biological Sciences, University of Memphis, Memphis, TN, USA
| | - Quincy A Quick
- Department of Biological Sciences, Tennessee State University, Nashville, TN, USA
| |
Collapse
|
31
|
Xue L, Jiang Y, Han T, Zhang N, Qin L, Xin H, Zhang Q. Comparative proteomic and metabolomic analysis reveal the antiosteoporotic molecular mechanism of icariin from Epimedium brevicornu maxim. JOURNAL OF ETHNOPHARMACOLOGY 2016; 192:370-381. [PMID: 27422162 DOI: 10.1016/j.jep.2016.07.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 07/10/2016] [Accepted: 07/11/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Icariin, a principal flavonoid glycoside of Epimedium brevicornu Maxim, has been widely proved to possess antiosteoporotic activity with promoting bone formation and decreasing bone resorption. However, the involving mechanisms remain unclear. AIM OF THE STUDY To clear a global insight of signal pathways involved in anti-osteoporotic mechanism of icariin at proteins and metabolites level by integrating the proteomics and NMR metabonomics, in a systems biology approach. MATERIAL AND METHODS Mice were divided into sham, OVX model and icariin-treated OVX group, after 90 days treatment, difference gel electrophoresis combined with MALDI-TOF/TOF proteomics analysis on bone femur and serum metabolomics were carried out for monitor intracellular processes and elucidate anti-osteoporotic mechanism of icariin. Osteoblast and osteoclast were applied to evaluate the potential signal pathways. RESULTS Twenty three proteins in bone femur, and 8 metabolites in serum, were significantly altered and identified, involving in bone remodeling, energy metabolism, cytoskeleton, lipid metabolism, MAPK signaling, Ca2+ signaling et, al. Furthermore, animal experiment show icariin could enhance the BMD and BMC, decrease CTX-I level in ovariectomized mice. The mitochondrial membrane potential and the intracellular ATP levels were increased significantly, and the cytoskeleton were improved in icariin-treatment osteoblast and osteoclast. Icariin also increased mRNA expression of Runx2 and osterix of OB, decreased CTR and CAII mRNA expression and protein expression of P38 and JNK. However, icariin did not reveal any inhibition of the collagenolytic activity of cathepsin K, mRNA expression of MMP-9 and protein expression of ERK in osteoclast. CONCLUSION we consider icariin as multi-targeting compounds for treating with osteoporosis, involve initiating osteoblastogenesis, inhibiting adipogenesis, and preventing osteoclast differentiation.
Collapse
MESH Headings
- Adipogenesis/drug effects
- Animals
- Biomarkers/blood
- Bone Density/drug effects
- Bone Density Conservation Agents/isolation & purification
- Bone Density Conservation Agents/pharmacology
- Bone Remodeling/drug effects
- Cell Differentiation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Electrophoresis, Gel, Two-Dimensional
- Epimedium/chemistry
- Female
- Femur/drug effects
- Femur/metabolism
- Flavonoids/isolation & purification
- Flavonoids/pharmacology
- Gene Expression Regulation/drug effects
- Metabolomics/methods
- Mice, Inbred ICR
- Osteoblasts/drug effects
- Osteoblasts/metabolism
- Osteoclasts/drug effects
- Osteoclasts/metabolism
- Osteoporosis/blood
- Osteoporosis/drug therapy
- Osteoporosis/genetics
- Phytotherapy
- Plant Extracts/isolation & purification
- Plant Extracts/pharmacology
- Plants, Medicinal
- Proteomics/methods
- Proton Magnetic Resonance Spectroscopy
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction/drug effects
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
Collapse
Affiliation(s)
- Liming Xue
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai 200433, China; Institute of Chemical Toxicity, Shanghai Municipal Center for Disease Control and Prevention, Shanghai 200336, China; Department of Oral Biological and Medical Sciences, The University of British Columbia, Vancouver, BC, Canada V6T1Z3
| | - Yiping Jiang
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Ting Han
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Naidan Zhang
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Luping Qin
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Hailiang Xin
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Qiaoyan Zhang
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
32
|
Hu L, Su P, Li R, Yin C, Zhang Y, Shang P, Yang T, Qian A. Isoforms, structures, and functions of versatile spectraplakin MACF1. BMB Rep 2016; 49:37-44. [PMID: 26521939 PMCID: PMC4914211 DOI: 10.5483/bmbrep.2016.49.1.185] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Indexed: 11/20/2022] Open
Abstract
Spectraplakins are crucially important communicators, linking cytoskeletal components to each other and cellular junctions. Microtubule actin crosslinking factor 1 (MACF1), also known as actin crosslinking family 7 (ACF7), is a member of the spectraplakin family. It is expressed in numerous tissues and cells as one extensively studied spectraplakin. MACF1 has several isoforms with unique structures and well-known function to be able to crosslink F-actin and microtubules. MACF1 is one versatile spectraplakin with various functions in cell processes, embryo development, tissue-specific functions, and human diseases. The importance of MACF1 has become more apparent in recent years. Here, we summarize the current knowledge on the presence and function of MACF1 and provide perspectives on future research of MACF1 based on our studies and others. [BMB Reports 2016; 49(1): 37-44]
Collapse
Affiliation(s)
- Lifang Hu
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Peihong Su
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Runzhi Li
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Chong Yin
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Yan Zhang
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Peng Shang
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Tuanmin Yang
- Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P. R. China
| | - Airong Qian
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| |
Collapse
|
33
|
Adipose-derived stem cell exosomes alleviate pathology of amyotrophic lateral sclerosis in vitro. Biochem Biophys Res Commun 2016; 479:434-439. [PMID: 27641665 DOI: 10.1016/j.bbrc.2016.09.069] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 09/14/2016] [Indexed: 01/11/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a degenerative disorder that involves the death of motor neurons in the cortex, brain stem, and spinal cord. Adipose-derived stem cells (ADSCs) are considered as a perspective remedy for therapy of neurodegenerative diseases including ALS. Stem cells secrete various factors which can modulate a hostile environment, called paracrine effect. Exosomes are small extracellular vesicles containing cell derived factors and mediate paracrine effect of cells. Thus, exosomes from ADSCs (ADSC-exo) can be a potential candidate of therapeutic effects of stem cells. To investigate the effect of ADSC-exo on the cellular phenotypes of ALS, we used neuronal stem cells (NSCs), which can be differentiated into neuronal cells, isolated from wild type or G93A ALS mice model. ADSC-exo was treated to neuronal cells from G93A ALS mice model. Immunocytochemistry and dot-blot assay result showed that ADSC-exo alleviated aggregation of superoxide dismutase 1 (SOD1). Reduction of cytosolic SOD1 level by ADSC-exo was also confirmed by western blot. Mitochondria display various abnormalities in ALS and the decrease of phospho-CREB and PGC-1α were observed in the G93A cells. ADSC-exo treatment showed normalization of phospho-CREB/CREB ratio and PGC-1α expression level. Our results suggest that ADSC-exo modulates cellular phenotypes of ALS including SOD-1 aggregation and mitochondrial dysfunction, and can be a therapeutic candidate for ALS.
Collapse
|