1
|
Ke D, Gao T, Dai H, Xu J, Ke T. RANKL promotes MT2 degradation and ROS production in osteoclast precursors through Beclin1-dependent autophagy. Differentiation 2025; 143:100863. [PMID: 40267773 DOI: 10.1016/j.diff.2025.100863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
ROS produced under oxidative stress are crucial for osteoclast differentiation. Metallothionein (MT) is a ROS-scavenging molecule. As a member of MT family, MT2 can clear ROS in osteoclast precursors (OCPs) and contributes to osteoclast differentiation. RANKL can promote OCP autophagy. Given the molecular-degrading effect of autophagy, the relationship between RANKL-dependent autophagy, MT2 and ROS during osteoclast differentiation is worth exploring. We depended in vitro RANKL administration and RANKL-overexpressing (Tg-RANKL) mice to observe the effects of RANKL on ROS production, MT2 protein expression, Beclin1 expression and autophagic activity in OCPs. Spautin1 was used to investigate the relationship between Beclin1-dependent autophagy and RANKL-regulated MT2 expression. Osteoclast-targeting MT2-cDNA-AAVs were applied to assess the therapeutic effect of MT2 on Tg-RANKL-related bone loss. The results showed that RANKL promoted ROS production but reduced MT2 protein expression in OCPs. RANKL also enhanced Beclin1 expression and LC3-puncta abundance. Decreased Beclin1 expression with spautin1 blocked RANKL-increased ROS production and osteoclast differentiation and recovered RANKL-decreased MT2 expression. MT2 selective overexpression with CD11b-promoter-MT2-cDNA-AAVs attenuated ROS production and osteoclastogenesis in Tg-RANKL mice and improved bone loss. Overall, RANKL can reduce MT2 protein expression through Beclin1-dependent autophagy, thereby promoting ROS production and osteoclast differentiation; this suggests that MT2-overexpressing small molecule drugs have the potential to treat RANKL-related bone loss.
Collapse
Affiliation(s)
- Dianshan Ke
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China
| | - Tingwei Gao
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China
| | - Hanhao Dai
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China
| | - Jie Xu
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China.
| | - Tie Ke
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China.
| |
Collapse
|
2
|
Chatterjee S, Sil PC. Mechanistic Insights into Toxicity of Titanium Dioxide Nanoparticles at the Micro- and Macro-levels. Chem Res Toxicol 2024; 37:1612-1633. [PMID: 39324438 DOI: 10.1021/acs.chemrestox.4c00235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Titanium oxide nanoparticles (TiO2 NPs) have been regarded as a legacy nanomaterial due to their widespread usage across multiple fields. The TiO2 NPs have been and are still extensively used as a food and cosmetic additive and in wastewater and sewage treatment, paints, and industrial catalysis as ultrafine TiO2. Recent developments in nanotechnology have catapulted it into a potent antibacterial and anticancer agent due to its excellent photocatalytic potential that generates substantial amounts of highly reactive oxygen radicals. The method of production, surface modifications, and especially size impact its toxicity in biological systems. The anatase form of TiO2 (<30 nm) has been found to exert better and more potent cytotoxicity in bacteria as well as cancer cells than other forms. However, owing to the very small size, anatase particles are able to penetrate deep tissue easily; hence, they have also been implicated in inflammatory reactions and even as a potent oncogenic substance. Additionally, TiO2 NPs have been investigated to assess their toxicity to large-scale ecosystems owing to their excellent reactive oxygen species (ROS)-generating potential compounded with widespread usage over decades. This review discusses in detail the mechanisms by which TiO2 NPs induce toxic effects on microorganisms, including bacteria and fungi, as well as in cancer cells. It also attempts to shed light on how and why it is so prevalent in our lives and by what mechanisms it could potentially affect the environment on a larger scale.
Collapse
Affiliation(s)
- Sharmistha Chatterjee
- Division of Molecular Medicine, Bose Institute, P 1/12, CIT Scheme VIIM, Kankurgachi, Kolkata-700054, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P 1/12, CIT Scheme VIIM, Kankurgachi, Kolkata-700054, India
| |
Collapse
|
3
|
Zhang N, Wang L, Ye X. Pdk3's role in RANKL-induced osteoclast differentiation: insights from a bone marrow macrophage model. PeerJ 2024; 12:e18222. [PMID: 39399421 PMCID: PMC11470767 DOI: 10.7717/peerj.18222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Background Osteoporosis (OP) is a chronic disease characterized by decreased bone mass, loss of skeletal structural integrity and increased susceptibility to fracture. Available studies have shown that the pyruvate dehydrogenase kinase (PDK) family is associated with osteoclastogenesis and bone loss, but the specific role of Pdk3 in bone pathology has not been systematically investigated. Methods A cell OP model was established in receptor activator for nuclear factor-κB Ligand (RANKL)-induced bone marrow macrophages (BMMs). Hereafter, the expression levels of Pdk3 and osteoclastogenesis feature genes including nuclear factor of activated T cells 1 (Nfatc1), Cathepsin K (Ctsk), osteoclast associated Ig-like receptor (Oscar) in BMMs-derived osteoclasts were examined based on real-time quantitative PCR and western blotting methods. Further, the phosphorylation of ERK, P65 and JAK/STAT and their correlation was Pdk3 was gauged. In particular, changes in the activity of these signaling pathways were observed by silencing experiments of the Pdk3 gene (using small interfering RNA). Finally, the effects of Pdk3 gene silencing on signaling pathway activity, osteoclastogenesis, and related inflammatory and apoptotic indicators were observed by transfection with PDK3-specific siRNA. Results Following RANKL exposure, the levels of Pdk3 and osteoclastogenesis feature genes were all elevated, and a positive correlation between Pdk3 and osteoclastogenesis feature genes was seen. Meanwhile, ERK, P65 and JAK/STAT phosphorylation was increased by RANKL, and Pdk3 was confirmed to be positively correlated with the phosphorylation of ERK, P65 and JAK/STAT. Additionally, in RANKL-exposed osteoclasts, Pdk3 knockdown diminished the phosphorylation of ERK, P65 and JAK/STAT, reduced the expressions of osteoclastogenesis feature genes. Importantly, knockdown of Pdk3 also reduced the expression of inflammatory cytokines and resulted in elevated levels of Bax and Casp3 expression, as well as downregulation of Bcl2 expression. Conclusion This study reveals for the first time the role of Pdk3 in RANKL-induced osteoclastogenesis and OP. These findings provide a foundation for future studies on the role of Pdk3 in other bone diseases and provide new ideas for the development of OP therapeutics targeting Pdk3.
Collapse
Affiliation(s)
- Nan Zhang
- College of Physical Education, Anhui Normal University, Wuhu, China
| | - Lingting Wang
- Spinal Surgery, The First Affiliated Hospital of Wannan Medical Collage, Wuhu, China
| | - Xuxin Ye
- Office of Hospital Admission and Discharge, The First Affiliated Hospital of Wannan Medical Collage, Wuhu, China
| |
Collapse
|
4
|
Zhang H, Ge G, Zhang W, Sun H, Liang X, Xia Y, Du J, Wu Z, Bai J, Yang H, Yang X, Zhou J, Xu Y, Geng D. PP2Ac Regulates Autophagy via Mediating mTORC1 and ULK1 During Osteoclastogenesis in the Subchondral Bone of Osteoarthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404080. [PMID: 39041921 PMCID: PMC11423161 DOI: 10.1002/advs.202404080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/02/2024] [Indexed: 07/24/2024]
Abstract
The molecular mechanism underlying abnormal osteoclastogenesis triggering subchondral bone remodeling in osteoarthritis (OA) is still unclear. Here, single-cell and bulk transcriptomics sequencing analyses are performed on GEO datasets to identify key molecules and validate them using knee joint tissues from OA patients and rat OA models. It is found that the catalytic subunit of protein phosphatase 2A (PP2Ac) is highly expressed during osteoclastogenesis in the early stage of OA and is correlated with autophagy. Knockdown or inhibition of PP2Ac weakened autophagy during osteoclastogenesis. Furthermore, the ULK1 expression of the downstream genes is significantly increased when PP2Ac is knocked down. PP2Ac-mediated autophagy is dependent on ULK1 phosphorylation activity during osteoclastogenesis, which is associated with enhanced dephosphorylation of ULK1 Ser637 residue regulating at the post-translational level. Additionally, mTORC1 inhibition facilitated the expression level of PP2Ac during osteoclastogenesis. In animal OA models, decreasing the expression of PP2Ac ameliorated early OA progression. The findings suggest that PP2Ac is also a promising therapeutic target in early OA.
Collapse
Affiliation(s)
- Haifeng Zhang
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Department of Orthopaedic SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Gaoran Ge
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Wei Zhang
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Houyi Sun
- Department of OrthopedicsQilu Hospital of Shandong UniversityJinanShandong250063China
| | - Xiaolong Liang
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Yu Xia
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Jiacheng Du
- Department of Biochemistry and Molecular BiologyJeonbuk National University Medical SchoolJeonjuJeonbuk54896South Korea
| | - Zerui Wu
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Department of OrthopedicsChangshu Hospital Affiliated to Soochow UniversityChangshuJiangsu215501China
| | - Jiaxiang Bai
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Department of Orthopedicsthe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui234000China
| | - Huilin Yang
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Xing Yang
- Orthopedics and Sports Medicine CenterSuzhou Municipal HospitalNanjing Medical University Affiliated Suzhou Hospital242, Guangji RoadSuzhouJiangsu215008China
| | - Jun Zhou
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Yaozeng Xu
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| | - Dechun Geng
- Department of Orthopedics Surgerythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
| |
Collapse
|
5
|
Wang H, Yuan T, Wang Y, Liu C, Li D, Li Z, Sun S. Osteoclasts and osteoarthritis: Novel intervention targets and therapeutic potentials during aging. Aging Cell 2024; 23:e14092. [PMID: 38287696 PMCID: PMC11019147 DOI: 10.1111/acel.14092] [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: 10/17/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024] Open
Abstract
Osteoarthritis (OA), a chronic degenerative joint disease, is highly prevalent among the aging population, and often leads to joint pain, disability, and a diminished quality of life. Although considerable research has been conducted, the precise molecular mechanisms propelling OA pathogenesis continue to be elusive, thereby impeding the development of effective therapeutics. Notably, recent studies have revealed subchondral bone lesions precede cartilage degeneration in the early stage of OA. This development is marked by escalated osteoclast-mediated bone resorption, subsequent imbalances in bone metabolism, accelerated bone turnover, and a decrease in bone volume, thereby contributing significantly to the pathological changes. While the role of aging hallmarks in OA has been extensively elucidated from the perspective of chondrocytes, their connection with osteoclasts is not yet fully understood. There is compelling evidence to suggest that age-related abnormalities such as epigenetic alterations, proteostasis network disruption, cellular senescence, and mitochondrial dysfunction, can stimulate osteoclast activity. This review intends to systematically discuss how aging hallmarks contribute to OA pathogenesis, placing particular emphasis on the age-induced shifts in osteoclast activity. It also aims to stimulate future studies probing into the pathological mechanisms and therapeutic approaches targeting osteoclasts in OA during aging.
Collapse
Affiliation(s)
- Haojue Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Tao Yuan
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Yi Wang
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Changxing Liu
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Dengju Li
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Ziqing Li
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| |
Collapse
|
6
|
Wang X, Zhu W, Chen H, Li X, Zheng W, Zhang Y, Fan N, Chen X, Wang G. JNK signaling mediates acute rejection via activating autophagy of CD8 + T cells after liver transplantation in rats. Front Immunol 2024; 15:1359859. [PMID: 38562941 PMCID: PMC10982410 DOI: 10.3389/fimmu.2024.1359859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Background Acute rejection (AR) after liver transplantation (LT) remains an important factor affecting the prognosis of patients. CD8+ T cells are considered to be important regulatory T lymphocytes involved in AR after LT. Our previous study confirmed that autophagy mediated AR by promoting activation and proliferation of CD8+ T cells. However, the underlying mechanisms regulating autophagy in CD8+ T cells during AR remain unclear. Methods Human liver biopsy specimens of AR after orthotopic LT were collected to assess the relationship between JNK and CD8+ T cells autophagy. The effect of JNK inhibition on CD8+ T cells autophagy and its role in AR were further examined in rats. Besides, the underlying mechanisms how JNK regulated the autophagy of CD8+ T cells were further explored. Results The expression of JNK is positive correlated with the autophagy level of CD8+ T cells in AR patients. And similar findings were obtained in rats after LT. Further, JNK inhibitor remarkably inhibited the autophagy of CD8+ T cells in rat LT recipients. In addition, administration of JNK inhibitor significantly attenuated AR injury by promoting the apoptosis and downregulating the function of CD8+ T cells. Mechanistically, JNK may activate the autophagy of CD8+ T cells through upregulating BECN1 by inhibiting the formation of Bcl-2/BECN1 complex. Conclusion JNK signaling promoted CD8+ T cells autophagy to mediate AR after LT, providing a theoretical basis for finding new drug targets for the prevention and treatment of AR after LT.
Collapse
Affiliation(s)
- Xiaowen Wang
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenfeng Zhu
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Haoqi Chen
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xuejiao Li
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenjie Zheng
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuan Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ning Fan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaolong Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Genshu Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
7
|
Liu X, Ye Z, Rao D, Chen Q, Zhang Z. DUSP4 maintains the survival and LSD1 protein stability in esophageal squamous cell carcinoma cells by inhibiting JNK signaling-dependent autophagy. In Vitro Cell Dev Biol Anim 2024; 60:115-122. [PMID: 38286920 DOI: 10.1007/s11626-023-00845-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 12/13/2023] [Indexed: 01/31/2024]
Abstract
DUSP4 is a biomarker of esophageal squamous cell carcinoma (ESCC), which is responsible for the prognosis in ESCC. However, the underlying mechanism of DUSP4-regulated ESCC carcinogenesis is unknown. As a negative regulator of JNK, DUSP4 can inhibit autophagy, which contributes to tumorigenesis. This study aimed to explore the role of autophagy in DUSP4-regulated ESCC carcinogenesis. Our results showed that DUSP4 overexpression inhibited autophagy and promoted LSD1 protein expression in ESCC cells, while DUSP4 silencing showed the opposite effects. However, DUSP4 overexpression and silencing did not affect LSD1 mRNA expression. But the regulatory ability of DUSP4 overexpression on autophagy, death level, and LSD1 protein was reversed by rapamycin. In addition, DUSP4 overexpression inhibited JNK and Bcl2 phosphorylation and the dissociation of Bcl2-Beclin1 complex, while DUSP4 silencing promoted JNK and Bcl2 phosphorylation. Moreover, the regulatory ability of DUSP4 overexpression on autophagy, death, and LSD1 protein was reversed by JNK activator anisomycin. The xenograft assays also showed that DUSP4 overexpression-promoted ESCC tumor growth in vivo and LC3II and LSD1 protein expression in tumor tissues were reversed by rapamycin or anisomycin. Overall, DUSP4 inhibits Bcl2-Beclin1-autophagy signal transduction through the negative regulation of JNK, thus suppressing autophagic death and the autophagic degradation of LSD1 in ESCC, by which DUSP4 promotes ESCC carcinogenesis.
Collapse
Affiliation(s)
- Xinxin Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
- Gannan Branch of National Clinical Research Center for Geriatrics Ganzhou 341000, Jiangxi, China
| | - Zhou Ye
- Department of Digestive, The 900Th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350001, Fujian, China
| | - Dingyu Rao
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, No. 3, Outangli, Xingannan Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Qianshun Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian, China.
| | - Zuxiong Zhang
- Department of Thoracic Surgery, First Affiliated Hospital of Gannan Medical University, No. 3, Outangli, Xingannan Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China.
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
8
|
Ke D, Xu H, Han J, Dai H, Wang X, Luo J, Yu Y, Xu J. Curcumin suppresses RANKL-induced osteoclast precursor autophagy in osteoclastogenesis by inhibiting RANK signaling and downstream JNK-BCL2-Beclin1 pathway. Biomed J 2024; 47:100605. [PMID: 37179010 PMCID: PMC10839592 DOI: 10.1016/j.bj.2023.100605] [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: 08/25/2022] [Revised: 01/30/2023] [Accepted: 05/08/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Curcumin ameliorates bone loss by inhibiting osteoclastogenesis. Curcumin inhibits RANKL-promoted autophagy in osteoclast precursors (OCPs), which mediates its anti-osteoclastogenic effect. But the role of RANKL signaling in curcumin-regulated OCP autophagy is unknown. This study aimed to explore the relationship between curcumin, RANKL signaling, and OCP autophagy during osteoclastogenesis. METHODS We investigated the role of curcumin in RANKL-related molecular signaling in OCPs, and identified the significance of RANK-TRAF6 signaling in curcumin-treated osteoclastogenesis and OCP autophagy using flow sorting and lentiviral transduction. Tg-hRANKL mice were used to observe the in vivo effects of curcumin on RANKL-regulated bone loss, osteoclastogenesis, and OCP autophagy. The significance of JNK-BCL2-Beclin1 pathway in curcumin-regulated OCP autophagy with RANKL was explored via rescue assays and BCL2 phosphorylation detection. RESULTS Curcumin inhibited RANKL-related molecular signaling in OCPs, and repressed osteoclast differentiation and autophagy in sorted RANK+ OCPs but did not affect those of RANK- OCPs. Curcumin-inhibited osteoclast differentiation and OCP autophagy were recovered by TRAF6 overexpression. But curcumin lost these effects under TRAF6 knockdown. Furthermore, curcumin prevented the decrease in bone mass and the increase in trabecular osteoclast formation and autophagy in RANK+ OCPs in Tg-hRANKL mice. Additionally, curcumin-inhibited OCP autophagy with RANKL was reversed by JNK activator anisomycin and TAT-Beclin1 overexpressing Beclin1. Curcumin inhibited BCL2 phosphorylation at Ser70 and enhanced protein interaction between BCL2 and Beclin1 in OCPs. CONCLUSIONS Curcumin suppresses RANKL-promoted OCP autophagy by inhibiting signaling pathway downstream of RANKL, contributing to its anti-osteoclastogenic effect. Moreover, JNK-BCL2-Beclin1 pathway plays an important role in curcumin-regulated OCP autophagy.
Collapse
Affiliation(s)
- Dianshan Ke
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, Fujian, China; Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Haoying Xu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Junyong Han
- Institute for Immunology, Fujian Academy of Medical Sciences, Fuzhou, Fujian, China
| | - Hanhao Dai
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, Fujian, China; Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Xinwen Wang
- Department of Orthopedics, Dongguan People's Hospital, Southern Medical University, Dongguan, Guangdong, China
| | - Jun Luo
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, Fujian, China; Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Yunlong Yu
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, Fujian, China; Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China.
| | - Jie Xu
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, Fujian, China; Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
9
|
Yu X, Wu Q, Ren Z, Chen B, Wang D, Yuan T, Ding H, Wang Y, Yuan G, Wang Y, Zhang L, Zhao J, Sun Z. Kaempferol attenuates wear particle-induced inflammatory osteolysis via JNK and p38-MAPK signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117019. [PMID: 37574017 DOI: 10.1016/j.jep.2023.117019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wear particle-induced inflammatory osteoclast activation is a master contributor to periprosthetic osteolysis, which can cause pathological bone loss and destruction. Hence, inhibiting inflammation and osteoclastogenesis is an important strategy for preventing wear particle-induced osteolysis. To date, there are no FDA-approved non-surgical pharmacotherapies for arresting periprosthetic osteolysis. Kaempferol (KAE), a natural flavonol abundant in many traditional Chinese herbal medicines, has been shown to have protective effects against inflammatory bone diseases such as rheumatoid arthritis, but no previous study has evaluated the effects of KAE on wear particle-induced osteolysis. AIM OF THE STUDY The study aimed to investigate the effects of KAE on wear particle-induced inflammatory osteolysis and osteoclast activation, and further explore the underlying mechanisms. MATERIALS AND METHODS TiAl6V4 metal particles (TiPs) were retrieved from the prosthesis of patients who underwent revision hip arthroplasty due to aseptic loosening. A mouse calvarial osteolysis model was used to investigate the effects of KAE on wear particle-induced inflammatory osteolysis in vivo. Primary bone marrow-derived macrophages (BMMs) were used to explore the effects of KAE on osteoclast differentiation and bone-resorbing activity as well as the underlying mechanisms in vitro. RESULTS In the present study, we found that KAE alleviated wear particle-induced inflammatory bone loss in vivo and inhibited osteoclast differentiation and function in vitro. Furthermore, we revealed that KAE exerted anti-osteoclastogenic effects by downregulating JNK and p38-MAPK signaling as well as the downstream NFATc1 expression. CONCLUSIONS KAE is an alternative therapeutic agent for preventing and treating periprosthetic osteolysis and aseptic loosening.
Collapse
Affiliation(s)
- Xin Yu
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Qi Wu
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China; Department of Vascular Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Zhengrong Ren
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, 210023, China
| | - Bin Chen
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Dongsheng Wang
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Tao Yuan
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Hao Ding
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Yang Wang
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Guodong Yuan
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Yuxiang Wang
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China
| | - Lei Zhang
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Jianning Zhao
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Zhongyang Sun
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School, Nanjing University, Nanjing, 210093, China; Department of Orthopedics, Air Force Hospital of Eastern Theater, Anhui Medical University, Nanjing, 210002, China.
| |
Collapse
|
10
|
Takahashi I, Watanabe Y, Sonoda H, Tsunoda D, Amano I, Koibuchi N, Iizuka H, Shimokawa N. Calcium sensing and signaling are impaired in the lumbar spine of a rat model of congenital kyphosis. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2023; 32:3403-3412. [PMID: 37555955 DOI: 10.1007/s00586-023-07877-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 07/07/2023] [Accepted: 07/28/2023] [Indexed: 08/10/2023]
Abstract
PURPOSE Kyphosis involves spines curving excessively backward beyond their physiological curvature. Although the normal structure of the spinal vertebrae is extremely important for maintaining posture and the normal function of the thoracic and abdominal organs, our knowledge concerning the pathogenesis of the disease is insufficient. We herein report that the downregulation of the calcium signaling pathway is involved in the pathogenesis of congenital kyphosis. METHODS The third to fifth lumbar spine segments, the kyphotic region of Ishibashi (IS) rats, which are used as a model of congenital kyphoscoliosis, were collected. A DNA microarray, quantitative PCR, Western blotting, and immunohistochemistry were used to measure the expression of genes and proteins related to intracellular calcium signaling. RESULTS We found that the expression of calcium-sensing receptor (CaSR) and transient receptor potential vanilloid 1 (Trpv1)-two receptors involved in the calcium signaling-was decreased in the lumbar spine of IS rats. We also observed that the number of CaSR-immunoreactive and Trpv1-immunoreactive cells in the lumbar spine of IS rats was lower than in wild-type rats. Furthermore, the expression of intracellular molecules downstream of these receptors, such as phosphorylated protein kinase C, c-Jun N-terminal kinase, and neural EGFL-like 1, was also reduced. In fact, the calcium content in the lumbar spine of IS rats was significantly lower than that in wild-type rats. CONCLUSION These results indicate that adequate calcium signaling is extremely important for the regulation of normal bone formation and may also be a key factor for understanding the pathogenesis of congenital kyphosis.
Collapse
Affiliation(s)
- Itsuki Takahashi
- Department of Nutrition, Takasaki University Graduate Scholl of Health and Welfare, 37-1 Nakaorui-Machi, Takasaki, Gunma, 370-0033, Japan
| | - Yusuke Watanabe
- Department of Nutrition, Takasaki University Graduate Scholl of Health and Welfare, 37-1 Nakaorui-Machi, Takasaki, Gunma, 370-0033, Japan
| | - Hiroyuki Sonoda
- Orthopaedic Surgery, Japanese Red Cross Maebashi Hospital, Maebashi, Gunma, Japan
| | - Daisuke Tsunoda
- Spine Surgery, Higashi-Maebashi Orthopaedic Hospital, Maebashi, Gunma, Japan
| | - Izuki Amano
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Haku Iizuka
- Orthopedic Surgery, Isesaki Municipal Hospital, Isesaki, Gunma, Japan.
| | - Noriaki Shimokawa
- Department of Nutrition, Takasaki University Graduate Scholl of Health and Welfare, 37-1 Nakaorui-Machi, Takasaki, Gunma, 370-0033, Japan.
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.
| |
Collapse
|
11
|
Arnst J, Jing Z, Cohen C, Ha SW, Viggeswarapu M, Beck GR. Bioactive silica nanoparticles target autophagy, NF-κB, and MAPK pathways to inhibit osteoclastogenesis. Biomaterials 2023; 301:122238. [PMID: 37441901 PMCID: PMC10530178 DOI: 10.1016/j.biomaterials.2023.122238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/28/2023] [Accepted: 07/02/2023] [Indexed: 07/15/2023]
Abstract
Spherical 50 nm silica-based nanoparticles (SiNPs) promote healthy bone homeostasis and maintenance by supporting bone forming osteoblast lineage cells while simultaneously inhibiting the differentiation of bone resorbing osteoclasts. Previous work demonstrated that an intraperitoneal injection of SiNPs in healthy mice - both young and old - increased bone density and quality, suggesting the possibility that SiNPs represent a dual action therapeutic. However, the underlying mechanisms governing the osteoclast response to SiNPs have yet to be fully explored and defined. Therefore, the goals of this study were to investigate the cellular and molecular mechanisms by which SiNPs inhibit osteoclastogenesis. SiNPs strongly inhibited RANKL-induced osteoclast differentiation within the first hours and concomitantly inhibited early transcriptional regulators such as Nfatc1. SiNPs simultaneously stimulated expression of autophagy related genes p62 and LC3β dependent on ERK1/2 signaling pathway. Intriguingly, SiNPs were found to stimulate autophagosome formation while inhibiting the autophagic flux necessary for RANKL-stimulated osteoclast differentiation, resulting in the inhibition of both the canonical and non-canonical NF-κB signaling pathways and stabilizing TRAF3. These results suggest a model in which SiNPs inhibit osteoclastogenesis by inhibiting the autophagic machinery and RANKL-dependent functionality. This mechanism of action defines a novel therapeutic strategy for inhibiting osteoclastogenesis.
Collapse
Affiliation(s)
- Jamie Arnst
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA
| | - Zhaocheng Jing
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA; The Second Hospital of Shandong University, Department of Orthopedics, Jinan, Shandong, 250033, China
| | - Cameron Cohen
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA
| | - Shin-Woo Ha
- Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA
| | - Manjula Viggeswarapu
- The Atlanta Department of Veterans Affairs Medical Center, Decatur, GA, 30033, USA
| | - George R Beck
- The Atlanta Department of Veterans Affairs Medical Center, Decatur, GA, 30033, USA; Emory University, Department of Medicine, Division of Endocrinology, Atlanta, GA, 30322, USA; The Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
12
|
Cai S, Chen Y, Chen J, Wei W, Pan J, Wu H. Rubiadin-1-methyl ether inhibits BECN1 transcription and Beclin1-dependent autophagy during osteoclastogenesis by inhibiting NF-κB p65 activation. Exp Biol Med (Maywood) 2023; 248:1518-1526. [PMID: 37750211 PMCID: PMC10666728 DOI: 10.1177/15353702231198071] [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: 04/19/2023] [Accepted: 06/07/2023] [Indexed: 09/27/2023] Open
Abstract
As an active substance isolated from the root of Morinda officinalis How., rubiadin-1-methyl ether (RBM), can improve osteoporosis due to its inhibition on osteoclastogenesis. Autophagy plays a key role in osteoclastogenesis. Our research aims to explore the relationship between RBM, autophagy, and osteoclastogenesis. Our results showed that RBM not only inhibited the differentiation level of osteoclasts and the proliferation ability of osteoclast precursors (OCPs), but also repressed the autophagic activity in OCPs (LC3 transformation and the number of autophagosomes observed by transmission electron microscopy). However, RBM-inhibited osteoclast differentiation and OCP autophagy (LC3 transformation and LC3-puncta formation) could be reversed by the application of TAT-Beclin1. Moreover, RBM administration reduced RANKL-induced p65 phosphorylation and p65 nuclear translocation in OCPs. In addition, the addition of RBM inhibited Beclin1 protein level and BECN1 (the gene form of Beclin1) mRNA level in OCPs increased by RANKL. Importantly, the reduction in the expression of BECN1 and Beclin1, LC3 transformation, and osteoclastic differentiation in OCPs caused by RBM were reversed by p65 overexpression. In conclusion, RBM may reduce the transcription of BECN1 by inhibiting the activation of nuclear factor kappa B (NF-κB) p65, thereby inhibiting Beclin1-dependent autophagy and RANKL-induced osteoclastogenesis.
Collapse
Affiliation(s)
- Suizhen Cai
- Health Examination Center, The Second People’s Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
| | - Yuyu Chen
- Department of Endocrinology, The Second People’s Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
| | - Jiawei Chen
- Health Examination Center, The Second People’s Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
| | - Wen Wei
- Health Examination Center, The Second People’s Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
| | - Jinquan Pan
- Health Examination Center, The Second People’s Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou 350003, China
| | - Haojie Wu
- Department of Endocrinology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361004, China
| |
Collapse
|
13
|
Xin SL, Pan XL, Xu XY, Yu YY. USP10 Alleviates Palmitic Acid-induced Steatosis through Autophagy in HepG2 Cells. J Clin Transl Hepatol 2023; 11:45-57. [PMID: 36406315 PMCID: PMC9647103 DOI: 10.14218/jcth.2022.00060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/03/2022] [Accepted: 03/21/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease caused by over-nutrition. Impaired autophagy is closely related to NAFLD progression. Recently, ubiquitin-specific peptidase-10 (USP10) was reported to ameliorate hepatic steatosis, but the underlying mechanism is still unclear. In view of the potential effects of USP10 on autophagy, we investigated whether USP10 alleviated steatosis through autophagy. METHODS HepG2 cells were treated with palmitic acid (PA) to model NAFLD in vitro. Lentivirus was used to regulate USP10 level in cells. Autophagic regulators were used to autophagic progression in cells. Western blotting, real-time fluorescence quantitative polymerase chain reaction, lipid drop staining and immunofluorescent staining were performed to determine the effect of USP10 on lipid autophagy. Student's t-test and Tukey's post hoc test were used to compare the means among groups. RESULTS PA induced cellular steatosis with dependance on autophagy. USP10 overexpression alleviated PA-induced steatosis, restored autophagic activity, promoted autophagic flux, including synthesis and degradation of autophagosomes, and lipid-targeted autophagy. In the presence of autophagy inhibitors, the protective effectiveness of USP10 on steatosis decreased. Furthermore, the specific inhibitor to C-jun N-terminal protein kinase-1 (JNK1), DB07268, abolished USP10-induced autophagy. However, during early stage inhibition of JNK1, compensatory expression of tuberous sclerosis complex-2 (TSC2) maintained autophagy. The degree of TSC2-to-JNK1 compensation was positively associated with USP10 level. Functionally, JNK1 and TSC2 were involved in the lipid-lowering effect of USP10. CONCLUSIONS USP10 alleviated hepatocellular steatosis in autophagy-dependent manner. JNK1/TSC2 signaling pathways were required for USP10-induced autophagy.
Collapse
Affiliation(s)
- Sheng-Liang Xin
- Department of Infectious Diseases, Peking University First Hospital, Beijing, China
| | - Xiao-Li Pan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiao-Yuan Xu
- Department of Infectious Diseases, Peking University First Hospital, Beijing, China
| | - Yan-Yan Yu
- Department of Infectious Diseases, Peking University First Hospital, Beijing, China
- Correspondence to: Yan-Yan Yu, Department of Infectious Diseases, Peking University First Hospital, Xishiku Street NO.8, Beijing 100034, China. ORCID: https://orcid.org/0000-0002-7557-1305. Tel: +86-10-66551066, Fax: +86-10-83572022, E-mail:
| |
Collapse
|
14
|
Zang L, Song Y, Tian Y, Hu N. TAT-Beclin 1 represses the carcinogenesis of DUSP4-positive PTC by enhancing autophagy. Mol Biol Rep 2023; 50:1425-1436. [PMID: 36474060 DOI: 10.1007/s11033-022-08109-2] [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: 09/20/2021] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND DUSP4 is a pro-tumorigenic molecule of papillary thyroid carcinoma (PTC). DUSP4 also exists as an autophagic regulator. Moreover, DUSP4, as a negative regulator of MAPK, can prevent Beclin 1 from participating in autophagic response. This study aimed to explore whether TAT-Beclin 1, a recombinant protein of Beclin 1, could inhibit the tumorigenesis of DUSP4-positive PTC by regulating autophagy. METHODS First, we divided PTC tissues into three groups according to DUSP4 expression levels by immunohistochemical analyses, and evaluated the relationship between autophagic molecules (Beclin 1 and LC3II) and DUSP4 using Western blotting assays. After overexpression of DUSP4 by lentiviral transduction, the in vitro and in vivo roles of TAT-Beclin 1 on DUSP4-overexpressed PTC cells were assessed (including autophagic activity, cell survival and function, and tumor growth). The roles of TAT-Beclin 1 in the survival of DUSP4-silenced PTC cells were also evaluated. RESULTS Our results showed that the expression levels of autophagic proteins decreased with the increase of DUSP4 expression in PTC tissues. In PTC cells, DUSP4 overexpression-inhibited autophagic activity (including Beclin 1 expression, LC3 conversion rate and LC3-puncta formation) and -promoted cell proliferation and migration were reversed by TAT-Beclin 1 administration. In vivo assays also showed that DUSP4-overexpressed PTC cells had stronger tumorigenic ability and weaker autophagic activity, which was blocked by TAT-Beclin 1 administration. CONCLUSION TAT-Beclin 1, as an autophagic promoter, could repress the carcinogenesis of DUSP4-positive PTC, which implies that the use of TAT-Beclin 1 for the PTC patients' treatment might be determined according to the DUSP4 level in their tumors.
Collapse
Affiliation(s)
- Leilei Zang
- Department 5 of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050005, Hebei, China
| | - Yanmei Song
- Department of Infection Management/Public Health, Hebei People's Hospital, Shijiazhuang, 050057, Hebei, China
| | - Yanhua Tian
- Department 2 of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050005, Hebei, China
| | - Ning Hu
- Department 4 of General Surgery, The Second Hospital of Hebei Medical University, No.215 Heping West Road, Xinhua District, Shijiazhuang, 050005, Hebei, China.
| |
Collapse
|
15
|
Li Z, Li D, Su H, Xue H, Tan G, Xu Z. Autophagy: An important target for natural products in the treatment of bone metabolic diseases. Front Pharmacol 2022; 13:999017. [PMID: 36467069 PMCID: PMC9716086 DOI: 10.3389/fphar.2022.999017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/08/2022] [Indexed: 12/28/2024] Open
Abstract
Bone homeostasis depends on a precise dynamic balance between bone resorption and bone formation, involving a series of complex and highly regulated steps. Any imbalance in this process can cause disturbances in bone metabolism and lead to the development of many associated bone diseases. Autophagy, one of the fundamental pathways for the degradation and recycling of proteins and organelles, is a fundamental process that regulates cellular and organismal homeostasis. Importantly, basic levels of autophagy are present in all types of bone-associated cells. Due to the cyclic nature of autophagy and the ongoing bone metabolism processes, autophagy is considered a new participant in bone maintenance. Novel therapeutic targets have emerged as a result of new mechanisms, and bone metabolism can be controlled by interfering with autophagy by focusing on certain regulatory molecules in autophagy. In parallel, several studies have reported that various natural products exhibit a good potential to mediate autophagy for the treatment of metabolic bone diseases. Therefore, we briefly described the process of autophagy, emphasizing its function in different cell types involved in bone development and metabolism (including bone marrow mesenchymal stem cells, osteoblasts, osteocytes, chondrocytes, and osteoclasts), and also summarized research advances in natural product-mediated autophagy for the treatment of metabolic bone disease caused by dysfunction of these cells (including osteoporosis, rheumatoid joints, osteoarthritis, fracture nonunion/delayed union). The objective of the study was to identify the function that autophagy serves in metabolic bone disease and the effects, potential, and challenges of natural products for the treatment of these diseases by targeting autophagy.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hui Su
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haipeng Xue
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guoqing Tan
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhanwang Xu
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
16
|
A Review of Signaling Transduction Mechanisms in Osteoclastogenesis Regulation by Autophagy, Inflammation, and Immunity. Int J Mol Sci 2022; 23:ijms23179846. [PMID: 36077242 PMCID: PMC9456406 DOI: 10.3390/ijms23179846] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Osteoclastogenesis is an ongoing rigorous course that includes osteoclast precursors fusion and bone resorption executed by degradative enzymes. Osteoclastogenesis is controlled by endogenous signaling and/or regulators or affected by exogenous conditions and can also be controlled both internally and externally. More evidence indicates that autophagy, inflammation, and immunity are closely related to osteoclastogenesis and involve multiple intracellular organelles (e.g., lysosomes and autophagosomes) and certain inflammatory or immunological factors. Based on the literature on osteoclastogenesis induced by different regulatory aspects, emerging basic cross-studies have reported the emerging disquisitive orientation for osteoclast differentiation and function. In this review, we summarize the partial potential therapeutic targets for osteoclast differentiation and function, including the signaling pathways and various cellular processes.
Collapse
|
17
|
Kang KA, Yao CW, Piao MJ, Zhen AX, Fernando PDSM, Herath HMUL, Song SE, Cho SJ, Hyun JW. Anticolon Cancer Effect of Korean Red Ginseng via Autophagy- and Apoptosis-Mediated Cell Death. Nutrients 2022; 14:nu14173558. [PMID: 36079818 PMCID: PMC9460327 DOI: 10.3390/nu14173558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/13/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
Ginseng (Panax ginseng Meyer) has been used in East Asian traditional medicine for a long time. Korean red ginseng (KRG) is effective against several disorders, including cancer. The cytotoxic effects of KRG extract in terms of autophagy- and apoptosis-mediated cell death and its mechanisms were investigated using human colorectal cancer lines. KRG induced autophagy-mediated cell death with enhanced expression of Atg5, Beclin-1, and LC3, and formed characteristic vacuoles in HCT-116 and SNU-1033 cells. An autophagy inhibitor prevented cell death induced by KRG. KRG generated mitochondrial reactive oxygen species (ROS); antioxidant countered this effect and decreased autophagy. KRG caused apoptotic cell death by increasing apoptotic cells and sub-G1 cells, and by activating caspases. A caspase inhibitor suppressed cell death induced by KRG. KRG increased phospho-Bcl-2 expression, but decreased Bcl-2 expression. Moreover, interaction of Bcl-2 with Beclin-1 was attenuated by KRG. Ginsenoside Rg2 was the most effective ginsenoside responsible for KRG-induced autophagy- and apoptosis-mediated cell death. KRG induced autophagy- and apoptosis-mediated cell death via mitochondrial ROS generation, and thus its administration may inhibit colon carcinogenesis.
Collapse
Affiliation(s)
- Kyoung Ah Kang
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Korea
| | - Cheng Wen Yao
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Korea
| | - Mei Jing Piao
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Korea
| | - Ao Xuan Zhen
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Korea
| | | | | | - Seung Eun Song
- Department of Anesthesiology, Jeju National University Hospital, College of Medicine, Jeju National University, Jeju 63241, Korea
| | - Suk Ju Cho
- Department of Anesthesiology, Jeju National University Hospital, College of Medicine, Jeju National University, Jeju 63241, Korea
- Correspondence: (S.J.C.); (J.W.H.); Tel.: +82-64-717-2062 (S.J.C.); +82-64-754-3838 (J.W.H.)
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Korea
- Correspondence: (S.J.C.); (J.W.H.); Tel.: +82-64-717-2062 (S.J.C.); +82-64-754-3838 (J.W.H.)
| |
Collapse
|
18
|
Jiang C, Wang Y, Zhang M, Xu J. Cholesterol inhibits autophagy in RANKL-induced osteoclast differentiation through activating the PI3K/AKT/mTOR signaling pathway. Mol Biol Rep 2022; 49:9217-9229. [PMID: 35881223 DOI: 10.1007/s11033-022-07747-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 06/23/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND A dysregulated balance between bone formation and bone resorption controlled by osteoblast and osteoclast will lead to osteoporosis. Cholesterol (CHO) is a crucial factor leading to osteoporosis, and autophagy appears to involve it. Therefore, we aimed to study the molecular mechanism of autophagy in CHO-induced osteoclasts differentiation. METHODS Nuclear factor-κ B ligand as a receptor activator was used to induce osteoclasts differentiation of murine macrophage RAW264.7 treated with CHO, PI3-kinase inhibitor (LY294002), and Rapamycin (RAPA), respectively. Western blot assay was used to detect the expression of TRAP/ACP5 and the proteins involved in autophagy and the PI3K/AKT/mTOR signaling pathway. In addition, TRAP staining, bone resorption assay, and F-actin immunofluorescence were performed to evaluate the ability of osteoclast formation. Transmission electron microscopy and immunofluorescence were also executed to observed the expression of LC3B, and autophagosome. RESULTS When RAW264.7 was treated with 20 μg/mL CHO for 5 consecutive days, It exhibited the optimal osteoclast activity. In addition, CHO could inhibit autophagy and activate the PI3K/AKT/mTOR signaling pathway. Moreover, the effects of CHO on osteoclast differentiation and autophagy could partially be reversed by LY294002 and RAPA. CONCLUSION Therefore, our results demonstrated that CHO could inhibit autophagy during osteoclast differentiation by activating the PI3K/AKT/mTOR signaling pathway. These findings provided important theoretical basis for CHO in bone resorption and formation.
Collapse
Affiliation(s)
- Chunyan Jiang
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.,Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, Shandong, China.,Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, 250021, Shandong, China.,Department of Endocrinology, People's Hospital of Linyi, Linyi, Shandong, China
| | - Yan Wang
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.,Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, Shandong, China.,Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, 250021, Shandong, China
| | - Mengqi Zhang
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.,Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, Shandong, China.,Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, 250021, Shandong, China
| | - Jin Xu
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China. .,Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China. .,Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, Shandong, China. .,Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, 250021, Shandong, China.
| |
Collapse
|
19
|
Wang P, Pan Y, Yang C, Zhang L, Zhao Z, Ye K, Li L, Xia S, Lu X, Shi H, Li W, Yin M. TNFα activation and TGFβ blockage act synergistically for smooth muscle cell calcification in patients with venous thrombosis via TGFβ/ERK pathway. J Cell Mol Med 2022; 26:4479-4491. [PMID: 35808901 PMCID: PMC9357635 DOI: 10.1111/jcmm.17472] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/12/2022] [Accepted: 06/16/2022] [Indexed: 11/26/2022] Open
Abstract
Venous calcification has been observed in post‐thrombotic syndrome (PTS) patients; yet, the cell types and possible mechanisms regulating this process are still unclear. We evaluated the calcium deposition within the venous wall, the cell type involved in the calcified remodelling of the venous wall after thrombosis and explored possible mechanisms in vitro. Calcium deposition was found in human specimens of superficial thrombotic veins and was co‐localized with VSMCs markers αSMA and TAGLN (also known as SM22α). Besides, the expression of osteogenesis‐related genes was dramatically changed in superficial thrombotic veins. Moreover, the inhibition of the TGFβ signalling pathway after TNFα treatment effectively induced the expression of osteogenic phenotype markers, the calcium salt deposits and the obvious phosphorylation of ERK1/2 and JNK2 in the VSMCs calcification model. Supplementing TGFβ2 or blocking the activation of the ERK/MAPK signalling pathway prevented the transformation of VSMCs into osteoblast‐like cells in vitro. Taken together, VSMCs have an important role in venous calcification after thrombosis. Supplementing TGFβ2 or inhibiting the ERK/MAPK signalling pathway can reduce the appearance of VSMCs osteogenic phenotype. Our findings may present a novel therapeutic approach to prevent of vascular calcification after venous thrombosis.
Collapse
Affiliation(s)
- Penghui Wang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yiqing Pan
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenghao Yang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Linjie Zhang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zhen Zhao
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Kaichuang Ye
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lei Li
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shoubing Xia
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Vascular Center of Shanghai JiaoTong University, Shanghai, China
| | - Huihua Shi
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Weimin Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Minyi Yin
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Vascular Center of Shanghai JiaoTong University, Shanghai, China
| |
Collapse
|
20
|
Guo J, Du X, Li C. BAG family proteins contributes to autophagy-mediated multidrug resistance of tumor. Clin Transl Oncol 2022; 24:1492-1500. [PMID: 35278199 DOI: 10.1007/s12094-022-02819-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/22/2022] [Indexed: 12/13/2022]
Abstract
Multidrug resistance (MDR) is a significant cause of tumor treatment failure. Accumulating evidence suggests that autophagy plays a significant role in the development of MDR. Autophagy is a conserved mechanism that maintains tumor homeostasis by removing damaged mitochondria. However, the specific regulatory mechanism is unclear. Here, we summarize recent studies on the role of autophagy in the development of MDR and the initiation of mitophagy by Bcl-2-associated athanogene (BAG) family proteins. Additionally, this mini-review emphasizes the regulatory role of BAG family proteins, which maintain mitochondrial homeostasis by regulating the PINK1/Parkin pathway. Elucidation of the regulatory mechanisms of mitophagy may foster the development of clinical therapeutic strategies for MDR tumors.
Collapse
Affiliation(s)
- Jufang Guo
- Department of Obstetrics and Gynecology, Jinniu District Maternal and Child Health Hospital, Chengdu, China
| | - Xuelian Du
- Department of Obstetrics and Gynecology, Jinniu District Maternal and Child Health Hospital, Chengdu, China
| | - Chaolin Li
- Department of Obstetrics and Gynecology, Jinniu District Maternal and Child Health Hospital, Chengdu, China.
| |
Collapse
|
21
|
Zhang W, Jiang G, Zhou X, Huang L, Meng J, He B, Qi Y. α-Mangostin inhibits LPS-induced bone resorption by restricting osteoclastogenesis via NF-κB and MAPK signaling. Chin Med 2022; 17:34. [PMID: 35248101 PMCID: PMC8898470 DOI: 10.1186/s13020-022-00589-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/26/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Excessive osteoclast activation is an important cause of imbalanced bone remodeling that leads to pathological bone destruction. This is a clear feature of many osteolytic diseases such as rheumatoid arthritis, osteoporosis, and osteolysis around prostheses. Because many natural compounds have therapeutic potential for treating these diseases by suppressing osteoclast formation and function, we hypothesized that α-mangostin, a natural compound isolated from mangosteen, might be a promising treatment as it exhibits anti‐inflammatory, anticancer, and cardioprotective effects.
Methods
We evaluated the therapeutic effect of α-mangostin on the processes of osteoclast formation and bone resorption. The receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL) induces osteoclast formation in vitro, and potential pathways of α-mangostin to inhibit osteoclast differentiation and function were explored. A mouse model of lipopolysaccharide‐induced calvarial osteolysis was established. Subsequently, micro-computed tomography and histological assays were used to evaluate the effect of α-mangostin in preventing inflammatory osteolysis.
Results
We found that α-mangostin could inhibit RANKL-induced osteoclastogenesis and reduced osteoclast‐related gene expression in vitro. F-actin ring immunofluorescence and resorption pit assays indicated that α-mangostin also inhibited osteoclast functions. It achieved these effects by disrupting the activation of NF-κB/mitogen-activated protein kinase signaling pathways. Our in vivo data revealed that α-mangostin could protect mouse calvarial bone from osteolysis.
Conclusions
Our findings demonstrate that α-mangostin can inhibit osteoclastogenesis both in vitro and in vivo and may be a potential option for treating osteoclast-related diseases.
Collapse
|
22
|
Phosphorylation of BCL2 at the Ser70 site mediates RANKL-induced osteoclast precursor autophagy and osteoclastogenesis. Mol Med 2022; 28:22. [PMID: 35183115 PMCID: PMC8858497 DOI: 10.1186/s10020-022-00449-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/24/2022] [Indexed: 11/23/2022] Open
Abstract
Background Phosphorylation modification of BCL2 is involved in receptor activator of nuclear factor-κB ligand (RANKL)-induced autophagy of osteoclast precursors (OCPs) and osteoclastogenesis. As an antiapoptotic molecule, the role of BCL2 phosphorylation in osteoclastogenesis is unknown. This study aimed to explore how BCL2 phosphorylation at specific sites regulates osteoclastogenesis.
Methods We first examined the effects of RANKL on BCL2 phosphorylation at different sites (Ser70 and Ser87) in OCPs. In vivo, transgenic mice overexpressing RANKL (Tg-hRANKL mice) were used to observe the effects of RANKL on phosphorylated BCL2 at different sites in OCPs of trabecular bone. Subsequently, using site-directed mutagenesis, we observed the respective effect of BCL2 mutations at different phosphorylation sites in OCPs on osteoclastogenesis, apoptosis, autophagy and the affinity between BCL2 and Beclin1/BAX under RANKL intervention. Results RANKL promoted BCL2 phosphorylation at the Ser70 (S70) site, but not the Ser87 (S87) site, in OCPs. Moreover, Tg-hRANKL mice had stronger BCL2 phosphorylation capacity at S70, not S87, in the OCPs of trabecular bone than wild-type mice in the same nest. Furthermore, BCL2 mutation at S70, not S87, inhibited RANKL-induced osteoclast differentiation and bone resorption activity. In addition, BCL2 mutation at S70 promoted OCP apoptosis, while BCL2 mutation at S87 showed the opposite effect. Remarkably, the BCL2 mutation at S70, not S87, inhibited OCP autophagic activity. Furthermore, BCL2 mutation at S70 enhanced the coimmunoprecipitation of BCL2 and Beclin1, whereas BCL2 mutation at S87 enhanced the coimmunoprecipitation of BCL2 and BAX in OCPs. More importantly, OCP autophagy, osteoclast differentiation and resorption pits inhibited by BCL2 mutation at S70 could be reversed by Beclin1 upregulation with TAT-Beclin1. Conclusion RANKL activates OCP autophagy through BCL2 phosphorylation at S70, thereby promoting osteoclastogenesis, which indicates that the inactivation of BCL2 at S70 in OCPs may be a therapeutic strategy for pathological bone loss. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00449-w.
Collapse
|
23
|
Ramser A, Greene E, Alrubaye AA, Wideman R, Dridi S. Role of Autophagy Machinery Dysregulation in Bacterial Chondronecrosis with Osteomyelitis (BCO). Poult Sci 2022; 101:101750. [PMID: 35278754 PMCID: PMC8914211 DOI: 10.1016/j.psj.2022.101750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 12/27/2021] [Accepted: 01/23/2022] [Indexed: 12/22/2022] Open
Abstract
Autophagy is a cell survival and homeostasis mechanism involving lysosomal degradation of cellular components and foreign bodies. It plays a role in bone homeostasis, skeletal diseases, and bacterial infections as both a cell-survival or cell-death pathway. This study sought to determine if autophagy played a role in bacterial chondronecrosis with osteomyelitis (BCO). BCO is a prominent cause of lameness in modern broilers and results from bacterial infection of mechanically stressed leg bone growth plates. The protein and gene expression of key autophagy machinery was analyzed in both normal and BCO-affected broilers using real-time qPCR and immunoblot, respectively. Gene expression showed a significant downregulation of key target signatures involved in every stage of autophagy in BCO-affected bone, such as ATG13, SQSTM1 (p62), ATG9B, ATG16L, ATG12, LC3C, and RAB7A. Additionally, protein expression for LC3 was also significantly lower in BCO. An in vitro study using human fetal osteoblast cells challenged with BCO isolate, Staphylococcus agnetis 908, showed a similar dysregulation of autophagy machinery along with a significant decrease in cell viability. When autophagy was inhibited via 3-methyladenine or chloroquine, comparable decreases in cell viability were seen along with dysregulation of autophagy machinery. Together, these results are the first to implicate autophagy machinery dysregulation in the pathology of BCO.
Collapse
|
24
|
He Y, Wang W, Xu X, Yang B, Yu X, Wu Y, Wang J. Mettl3 inhibits the apoptosis and autophagy of chondrocytes in inflammation through mediating Bcl2 stability via Ythdf1-mediated m 6A modification. Bone 2022; 154:116182. [PMID: 34530171 DOI: 10.1016/j.bone.2021.116182] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/20/2021] [Accepted: 09/07/2021] [Indexed: 12/01/2022]
Abstract
N6-methyladenosine (m6A) methylation is one of the most common internal modifications in eukaryotic messenger RNA occurring on N6 nitrogen of adenosine. However, the roles of m6A in temporomandibular joint osteoarthritis (TMJ OA) are still elusive. Here, we investigate the function and mechanism of methyltransferase-like 3 (Mettl3) in chondrocytes in inflammation. We found that the expression of Mettl3 decreased both in vivo TMJ OA mice and in vitro inflammatory stimulation. Functionally, loss and gain studies illustrated that Mettl3 inhibited the apoptosis and autophagy of chondrocytes induced by TNF-α stimulation in vitro. Mettl3 inhibitor, S-adenosylhomocysteine (SAH) promoted the apoptosis and autophagy of chondrocytes with inflammation in vitro and aggravated the degeneration of chondrocytes and subchondral bone in monosodium iodoacetate (MIA) induced TMJ OA mice in vivo. Mechanistically, the bioinformatics analysis, m6A-RNA immunoprecipitation (MeRIP) and RNA immunoprecipitation (RIP) were used to identify that Bcl2 mRNA was the downstream target of Mettl3 for m6A modification. Furthermore, the results revealed that Yth m6A RNA binding protein 1 (Ythdf1) mediated the stability of Bcl2 mRNA catalyzed by Mettl3. Co-immunoprecipitation (Co-IP) showed that Bcl2 protein interacted with Beclin1 protein in chondrocytes induced by TNF-α stimulation. In conclusion, our findings identify that Mettl3 inhibits the apoptosis and autophagy of chondrocytes in inflammation through m6A/Ythdf1/Bcl2 signal axis which provides promising therapeutic strategy for TMJ OA.
Collapse
Affiliation(s)
- Ying He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Wei Wang
- Departments of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Xiaoxiao Xu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Beining Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Xijie Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Yanru Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Jiawei Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China.
| |
Collapse
|
25
|
ATG 4B Serves a Crucial Role in RCE-4-Induced Inhibition of the Bcl-2-Beclin 1 Complex in Cervical Cancer Ca Ski Cells. Int J Mol Sci 2021; 22:ijms222212302. [PMID: 34830185 PMCID: PMC8617943 DOI: 10.3390/ijms222212302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/31/2021] [Accepted: 11/08/2021] [Indexed: 12/27/2022] Open
Abstract
RCE-4, a steroidal saponin isolated from Reineckia carnea, has been studied previously and has exhibited promising anti-cervical cancer properties by inducing programmed cell death (PCD) of Ca Ski cells. Considering the cancer cells developed various pathways to evade chemotherapy-induced PCD, there is, therefore, an urgent need to further explore the potential mechanisms underlying its actions. The present study focused on targeting the Bcl-2–Beclin 1 complex, which is known as the key regulator of PCD, to deeply elucidate the molecular mechanism of RCE-4 against cervical cancer. The effects of RCE-4 on the Bcl-2–Beclin 1 complex were investigated by using the co-immunoprecipitation assay. In addition, autophagy-related genes (ATG) were also analyzed due to their special roles in PCD. The results demonstrated that RCE-4 inhibited the formation of the Bcl-2–Beclin 1 complex in Ca Ski cells via various pathways, and ATG 4B proteins involved in this process served as a key co-factor. Furthermore, based on the above, the sensitivity of RCE-4 to Ca Ski cells was significantly enhanced by inhibiting the expression of the ATG 4B by applying the ATG 4B siRNA plasmid.
Collapse
|
26
|
Ke D, Wang X, Lin Y, Wei S. Lactoferrin promotes the autophagy activity during osteoblast formation via BCL2-Beclin1 signaling. Mol Biol Rep 2021; 49:259-266. [PMID: 34716503 DOI: 10.1007/s11033-021-06866-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/20/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Lactoferrin, as the main component of milk, can maintain osteoblast formation, which is conducive to the prevention and treatment of osteoporosis. Lactoferrin also serves as an autophagy regulator, especially in osteoblasts. This study aimed to explore the significance of autophagy in osteoblast formation regulated by lactoferrin and the internal mechanism. METHODS AND RESULTS In this study, we firstly explored the roles of lactoferrin in the autophagy activity of primary osteoblasts (LC3 transformation rate, autophagosome formation). Subsequently, we further investigated the effects of lactoferrin on the BCL2 expression and BCL2-Beclin1 complex. Ultimately, the significance of BCL2 overexpression and Beclin1 silencing on lactoferrin-regulated osteoblast autophagy and osteogenic parameters (ALP activity and mRNA expression of PCNA, Col1, BGLAP and OPN) was observed by gene processing, respectively. Our results showed that lactoferrin enhanced the autophagy activity of osteoblasts. Importantly, lactoferrin inhibited BCL2 expression and the co-immunoprecipitation of BCL2 and Beclin1 in osteoblasts. Moreover, lactoferrin-promoted autophagy and osteogenic parameters was reversed by BCL2 overexpression or Beclin1 silencing in osteoblasts. CONCLUSIONS In conclusion, lactoferrin can inhibit BCL2 expression in osteoblasts, further enhancing Beclin1-dependent autophagy activation.
Collapse
Affiliation(s)
- Dianshan Ke
- Department of Orthopedics, The People's Hospital of JiangMen, No. 172 Gaodi Li, Pengjiang District, Jiangmen, 529000, Guangdong, China
- Academy of Orthopedics in Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Xinwen Wang
- Department of Orthopedics, The People's Hospital of JiangMen, No. 172 Gaodi Li, Pengjiang District, Jiangmen, 529000, Guangdong, China
| | - Yinquan Lin
- Department of Orthopedics, The People's Hospital of JiangMen, No. 172 Gaodi Li, Pengjiang District, Jiangmen, 529000, Guangdong, China.
| | - Shengwang Wei
- Department of Orthopedics, Liuzhou Workers Hospital, No.47, Zone 4, Hongyan Road, Liunan District, Liuzhou, 545005, Guangxi, China.
| |
Collapse
|
27
|
Yu C, Zhu Y, Lv X, Wang Y. 1α,25-(OH) 2-D 3 promotes the autophagy during osteoclastogenesis by enhancing RANKL-RANK-TRAF6 signaling. In Vitro Cell Dev Biol Anim 2021; 57:878-885. [PMID: 34780049 DOI: 10.1007/s11626-021-00632-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/30/2021] [Indexed: 11/25/2022]
Abstract
As the active form of vitamin D3, 1α,25-(OH)2-D3 promotes receptor activator for nuclear factor-κB ligand (RANKL)-induced autophagy in osteoclast precursors (OCPs). However, the relationship between 1α,25-(OH)2-D3 and RANKL signaling is still unknown. This study aimed to explore whether 1α,25-(OH)2-D3 regulates OCP autophagy and osteoclastogenesis through RANKL signaling. Our results showed that 1α,25-(OH)2-D3 directly decreased OCP autophagy while significantly enhancing the ability of RANKL to promote OCP autophagy. Moreover, 1α,25-(OH)2-D3 not only promoted the expression of key signaling proteins in OCPs induced by RANKL but also enhanced the coimmunoprecipitation levels of RANK and TRAF6. Notably, 1α,25-(OH)2-D3 significantly enhanced the autophagic activity and osteoclast differentiation of RANK-positive OCPs but did not affect the autophagic activity or osteoclast differentiation of RANK-negative OCPs. More importantly, 1α,25-(OH)2-D3 had no effect on autophagy or osteoclastogenesis in TRAF6-silenced OCPs. Overall, 1α,25-(OH)2-D3 could upregulate RANKL-RANK-TRAF6 signaling in OCPs, thereby promoting OCP autophagy and osteoclastogenesis.
Collapse
Affiliation(s)
- Chengjian Yu
- Department of Emergency, 900 Hospital of The Joint Logistics Team, Dongfang Hospital, Xiamen University, Fuzong Clinical College of Fujian Medical University, Fuzhou, 350025, Fujian, China
| | - Yunrong Zhu
- Department of Orthopedics, The Affiliated Jiangyin Hospital of Medical College of Southeast University, No. 163 Shoushan Road, Jiangyin, 214400, Jiangsu, China.
| | - Xiaofei Lv
- Department of Orthopedics, Yixin Shanjuan Orthopaedic Hospital, YiXing, 214000, Jiangsu, China
| | - Yabin Wang
- Department of Orthopedics, The Affiliated Jiangyin Hospital of Medical College of Southeast University, No. 163 Shoushan Road, Jiangyin, 214400, Jiangsu, China
| |
Collapse
|
28
|
Tong X, Min W, Li S, Chen M, Song R, Bian J, Gu J, Liu Z. Beclin 1 positively regulates osteoprotegerin-induced inhibition of osteoclastogenesis by increasing autophagy in vitro. Differentiation 2021; 121:35-43. [PMID: 34454349 DOI: 10.1016/j.diff.2021.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 08/20/2021] [Indexed: 12/28/2022]
Abstract
Osteoclastogenesis is induced by receptor activator of nuclear factor-κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF), and can be suppressed by osteoprotegerin (OPG). Beclin1 has a dual role in osteoclastogenesis. However, the role of Beclin1-mediated autophagy during OPG-induced inhibition of osteoclastogenesis remains unclear. Here, we found that Beclin1 and matrix metalloproteinase 9 (MMP-9) expression were increased during osteoclastogenesis. OPG (20, 40, and 80 ng/mL) decreased Src and MMP-9 expression, but augmented Beclin1 expression and fluorescence intensity. Similarly, treatment with the autophagy activator rapamycin increased Beclin1 expression during OPG-induced inhibition of osteoclastogenesis. Further, Beclin1 knockdown restored osteoclast numbers by reducing autophagy during OPG-induced inhibition of osteoclastogenesis. These results indicate that Beclin1 has a positive role during OPG-induced inhibition of osteoclastogenesis by regulating autophagy, which might provide a potential basis for osteoclastogenesis.
Collapse
Affiliation(s)
- Xishuai Tong
- Institutes of Agricultural Science and Technology Development, Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, Jiangsu, PR China; College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, Jiangsu, PR China; Center of Excellence for Vector-Borne Diseases, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, 66502, Kansas, USA
| | - Wenyan Min
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, PR China
| | - Saihui Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, PR China
| | - Miaomiao Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, PR China
| | - Ruilong Song
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, PR China
| | - Jianchun Bian
- Institutes of Agricultural Science and Technology Development, Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, Jiangsu, PR China; College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, Jiangsu, PR China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, PR China
| | - Zongping Liu
- Institutes of Agricultural Science and Technology Development, Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, Jiangsu, PR China; College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, PR China; Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, Jiangsu, PR China.
| |
Collapse
|
29
|
Li X, Xu J, Dai B, Wang X, Guo Q, Qin L. Targeting autophagy in osteoporosis: From pathophysiology to potential therapy. Ageing Res Rev 2020; 62:101098. [PMID: 32535273 DOI: 10.1016/j.arr.2020.101098] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/26/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022]
Abstract
Osteoporosis is a highly prevalent disorder characterized by the loss of bone mass and microarchitecture deterioration of bone tissue, attributed to various factors, including menopause (primary), aging (primary) and adverse effects of relevant medications (secondary). In recent decades, knowledge regarding the etiological mechanisms underpinning osteoporosis emphasizes that bone cellular homeostasis, including the maintenance of cell functions, differentiation, and the response to stress, is tightly regulated by autophagy, which is a cell survival mechanism for eliminating and recycling damaged proteins and organelles. With the important roles in the maintenance of cellular homeostasis and organ function, autophagy has emerged as a potential target for the prevention and treatment of osteoporosis. In this review, we update and discuss the pathophysiology of autophagy in normal bone cell life cycle and metabolism. Then, the alternations of autophagy in primary and secondary osteoporosis, and the accompanied pathological process are discussed. Finally, we discuss current strategies, limitations, and challenges involved in targeting relevant pathways and propose strategies by which such hurdles may be circumvented in the future for their translation into clinical validations and applications for the prevention and treatment of osteoporosis.
Collapse
|
30
|
Chen XD, Tan JL, Feng Y, Huang LJ, Zhang M, Cheng B. Autophagy in fate determination of mesenchymal stem cells and bone remodeling. World J Stem Cells 2020; 12:776-786. [PMID: 32952858 PMCID: PMC7477662 DOI: 10.4252/wjsc.v12.i8.776] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/17/2020] [Accepted: 06/20/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been widely exploited as promising candidates in clinical settings for bone repair and regeneration in view of their self-renewal capacity and multipotentiality. However, little is known about the mechanisms underlying their fate determination, which would illustrate their effectiveness in regenerative medicine. Recent evidence has shed light on a fundamental biological role of autophagy in the maintenance of the regenerative capability of MSCs and bone homeostasis. Autophagy has been implicated in provoking an immediately available cytoprotective mechanism in MSCs against stress, while dysfunction of autophagy impairs the function of MSCs, leading to imbalances of bone remodeling and a wide range of aging and degenerative bone diseases. This review aims to summarize the up-to-date knowledge about the effects of autophagy on MSC fate determination and its role as a stress adaptation response. Meanwhile, we highlight autophagy as a dynamic process and a double-edged sword to account for some discrepancies in the current research. We also discuss the contribution of autophagy to the regulation of bone cells and bone remodeling and emphasize its potential involvement in bone disease.
Collapse
Affiliation(s)
- Xiao-Dan Chen
- Hospital of Stomatology, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Stomatology; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong Province, China
| | - Jia-Li Tan
- Hospital of Stomatology, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Stomatology; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong Province, China
| | - Yi Feng
- Hospital of Stomatology, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Stomatology; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong Province, China
| | - Li-Jia Huang
- Hospital of Stomatology, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Stomatology; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong Province, China
| | - Mei Zhang
- Hospital of Stomatology, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Stomatology; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong Province, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Stomatology; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, Guangdong Province, China
| |
Collapse
|
31
|
Cheng L, Zhu Y, Ke D, Xie D. Oestrogen-activated autophagy has a negative effect on the anti-osteoclastogenic function of oestrogen. Cell Prolif 2020; 53:e12789. [PMID: 32157750 PMCID: PMC7162800 DOI: 10.1111/cpr.12789] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/10/2020] [Accepted: 02/16/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES Oestrogen is known to inhibit osteoclastogenesis, and numerous studies have identified it as an autophagic activator. To date, the role of oestrogen in the autophagy of osteoclast precursors (OCPs) during osteoclastogenesis remains unclear. This study aimed to determine the effect of autophagy regulated by the biologically active form of oestrogen (17β-estradiol) on osteoclastogenesis. MATERIALS AND METHODS After treatment with 17β-estradiol in OCPs (from bone marrow-derived macrophages, BMMs) and ovariectomy (OVX) mice, we measured the effect of 17β-estradiol on the autophagy of OCPs in vitro and in vivo. In addition, we studied the role of autophagy in the OCP proliferation, osteoclast differentiation and bone loss regulated by 17β-estradiol using autophagic inhibitor or knock-down of autophagic genes. RESULTS The results showed that direct administration of 17β-estradiol enhanced the autophagic response of OCPs. Interestingly, 17β-estradiol inhibited the stimulatory effect of receptor activator of nuclear factor-κB ligand (RANKL) on the autophagy and osteoclastogenesis of OCPs. Moreover, 17β-estradiol inhibited the downstream signalling of RANKL. Autophagic suppression by pharmacological inhibitors or gene silencing enhanced the inhibitory effect of 17β-estradiol on osteoclastogenesis. In vivo assays showed that the autophagic inhibitor 3-MA not only inhibited the autophagic activity of the OCPs in the trabecular bone of OVX mice but also enhanced the ability of 17β-estradiol to ameliorate bone loss. CONCLUSIONS In conclusion, our study showed that oestrogen directly enhanced the autophagy of OCPs, which inhibited its anti-osteoclastogenic effect. Drugs based on autophagic inhibition may enhance the efficacy of oestrogen on osteoporosis.
Collapse
Affiliation(s)
- Liang Cheng
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration DiseasesThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Yunrong Zhu
- Department of OrthopedicsThe Affiliated Jiangyin Hospital of Medical College of Southeast UniversityJiangyinChina
| | - Dianshan Ke
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration DiseasesThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Denghui Xie
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration DiseasesThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| |
Collapse
|
32
|
Hassanpour M, Rezaie J, Darabi M, Hiradfar A, Rahbarghazi R, Nouri M. Autophagy modulation altered differentiation capacity of CD146 + cells toward endothelial cells, pericytes, and cardiomyocytes. Stem Cell Res Ther 2020; 11:139. [PMID: 32216836 PMCID: PMC7099797 DOI: 10.1186/s13287-020-01656-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/11/2020] [Accepted: 03/18/2020] [Indexed: 12/15/2022] Open
Abstract
Background To date, many attempts are employed to increase the regenerative potential of stem cells. In this study, we evaluated the hypothesis of whether an autophagy modulation could alter differentiation potency of CD146+ cells into mature pericyte, endothelial, and cardiomyocyte lineage. Methods In this study, CD146+cells were enriched from the human bone marrow aspirates and trans-differentiated into mature endothelial cells, pericytes, and cardiomyocytes after exposure to autophagy stimulator (50-μM Met)/inhibitor (15-μM HCQ). The protein levels of autophagy proteins were monitored by western blotting. NO content was measured using the Griess assay. Using real-time PCR assay and western blotting, we monitored the lineage protein and gene levels. Pro-inflammatory cytokine and angiocrine factors were measured by ELISA. The fatty acid change was determined by gas chromatography. We also measured exosome secretion capacity by measuring AChE activity and real-time PCR assay. Result Data revealed the modulation of autophagy factors, Beclin-1, P62, and LC3 II/I ratio in differentiating CD146+ cells after exposure to Met and HCQ (p < 0.05). The inhibition of autophagy increased NO content compared to the Met-treated cells (p < 0.05). Real-time PCR analysis showed that the treatment of CD146+ cells with autophagy modulators altered the expression of VE-cadherin, cTnI, and α-SMA (p < 0.05). Met increased the expression of VE-cadherin, α-SMA, and cTnI compared to the HCQ-treated cells (p < 0.05) while western blotting revealed the protein synthesis of all lineage-specific proteins under the stimulation and inhibition of autophagy. None statistically significant differences were found in the levels of Tie-1, Tie-2, VEGFR-1, and VEGFR-2 after autophagy modulation. Fatty acid profile analysis revealed the increase of unsaturated fatty acids after exposure to HCQ (p < 0.05). The treatment of cells with HCQ increased the levels of TNF-α and IL-6 compared to the Met-treated cells. Data revealed the increase of exosome biogenesis and secretion to the supernatant in cells treated with HCQ compared to the Met groups (p < 0.05). Conclusions In summary, autophagy modulation could alter differentiation potency of CD146+cells which is important in cardiac regeneration.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756, Iran.,Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirataollah Hiradfar
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756, Iran. .,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756, Iran.
| |
Collapse
|
33
|
Lin S, Ke D, Lin Y, Fu X, Yu Y. Puerarin inhibits the migration of osteoclast precursors and osteoclastogenesis by inhibiting MCP-1 production. Biosci Biotechnol Biochem 2020; 84:1455-1459. [PMID: 32154764 DOI: 10.1080/09168451.2020.1738912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Puerarin inhibits osteoclastogenesis and cells migration. This study aims to explore whether puerarin prevents osteoclastogenesis by inhibiting osteoclast precursors (OCPs) migration. The results showed that puerarin reduced MCP-1 production in OCPs, while inhibiting OCPs migration based on MCP-1. Puerarin reversed MCP-1-promoted osteoclastogenesis. CCR2 overexpression didn't increase osteoclastogenesis with puerarin. Therefore, puerarin prevents OCPs migration by reducing MCP-1, whereby inhibiting osteoclastogenesis.
Collapse
Affiliation(s)
- Sanfu Lin
- Department of Orthopedics, The Second Affiliated Hospital of Fujian Medical University , Quanzhou, Fujian, China
| | - Dianshan Ke
- Department of Orthopedics, The People's Hospital of JiangMen , Jiangmen, Guangdong, China
| | - Yinquan Lin
- Department of Orthopedics, The People's Hospital of JiangMen , Jiangmen, Guangdong, China
| | - Xiaomin Fu
- Division of Metabolism and Endocrinology, John Hopkins University , Baltimore, MD, USA
| | - Yunlong Yu
- Department of Orthopedics, Fujian Provincial Hospital , Fuzhou, Fujian, China
| |
Collapse
|
34
|
Ke D, Wang Y, Yu Y, Wang Y, Zheng W, Fu X, Han J, Zhang G, Xu J. Curcumin-activated autophagy plays a negative role in its anti-osteoclastogenic effect. Mol Cell Endocrinol 2020; 500:110637. [PMID: 31678610 DOI: 10.1016/j.mce.2019.110637] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/27/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND/PURPOSE It remains unclear what role curcumin plays in the autophagy of osteoclast precursors (OCPs) during osteoclastogenesis, since some researchers found that curcumin has the ability to inhibit osteoclastogenesis. While others have considered it as an autophagy activator. This study aimed to determine the effect of curcumin-regulated autophagy on osteoclastogenesis. RESULTS The results revealed that direct administration of curcumin enhanced the OCP autophagy response in bone marrow-derived macrophages (BMMs). Curcumin could also abate RANKL's stimulatory effect on OCP autophagy and osteoclastogenesis. Autophagic suppression related to pharmacological inhibitors or gene silencing could further enhance the inhibitory effect of curcumin on osteoclastogenesis. As expected, curcumin ameliorated ovariectomy (OVX)-induced bone loss and its effect could be promoted by an autophagy inhibitor (chloroquine). CONCLUSIONS In conclusion, curcumin can directly enhance the autophagic activity of OCPs, which inhibits its anti-osteoclastogeneic effects. Autophagy inhibition-based drugs are expected to enhance curcumin's efficacy in treating osteoporosis.
Collapse
Affiliation(s)
- Dianshan Ke
- Department of Orthopedics, The People's Hospital of JiangMen, Jiangmen, 529000, Guangdong, China; Academy of Orthopedics in Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China.
| | - Yu Wang
- Department of Orthopaedics, Chifeng Hospital, Chifeng, 024000, Inner Mongolia, China
| | - Yunlong Yu
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China
| | - Yongxuan Wang
- Department of Endocrine, Sanming First Hosptial, The Affiliated Sanming First Hospital of Fujian Medical University, Sanming, 365000, Fujian, China
| | - Wang Zheng
- Center for Cancer and Immunology Research, Children's National Medical Center, Washington, 20010, DC, USA
| | - Xiaomin Fu
- Division of Metabolism and Endocrinology, John Hopkins University, Baltimore, 21218, Maryland, USA
| | - Junyong Han
- Institute for Immunology, Fujian Academy of Medical Sciences, Fuzhou, 350003, Fujian, China
| | - Guoyou Zhang
- Department of Orthopaedics, Tongliao City Hospital, Tongliao, 028000, Inner Mongolia, China
| | - Jie Xu
- Department of Orthopedics, Fujian Provincial Hospital, Fuzhou, 350003, Fujian, China.
| |
Collapse
|
35
|
Ke D, Zhu Y, Zheng W, Fu X, Chen J, Han J. Autophagy mediated by JNK1 resists apoptosis through TRAF3 degradation in osteoclastogenesis. Biochimie 2019; 167:217-227. [DOI: 10.1016/j.biochi.2019.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/17/2019] [Indexed: 12/31/2022]
|
36
|
Zhang G, Wang Y, Tang G, Ma Y. Puerarin inhibits the osteoclastogenesis by inhibiting RANKL-dependent and -independent autophagic responses. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:269. [PMID: 31615565 PMCID: PMC6794871 DOI: 10.1186/s12906-019-2691-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/20/2019] [Indexed: 01/13/2023]
Abstract
Background Puerarin exerts therapeutic effect on osteoporosis due to its inhibitory effect on the formation of osteoclasts. Puerarin is also widely established as an autophagy inhibitor. The study aimed to investigate the significance of autophagy in Puerarin-treated osteoclast formation. Methods Osteoclast precursors (OCPs) derived from bone marrow-derived macrophages (BMMs) were treated with Puerarin along with RANKL or without RANKL, and then the autophagic parameters of OCPs (including autophagic proteins, LC3 transformation, autophagosome or LC3-puncta) were observed through Western Blotting, Transmission Electron Microscopy and Immunofluorescence assays. Next, after using overexpression vectors of autophagic genes (Atg7, Atg5 and BECN1) to alter autophagy activity, OCP proliferation was measured by Ethynyl deoxyuridine (EdU) assays and Cell Counting Kit-8 (CCK-8) kit, and osteoclast differentiation was assessed by Tartrate-resistant acid phosphatase (TRAP) staining. Results The results showed that Puerarin could directly inhibit the autophagy and proliferation of OCPs. Importantly, overexpression of autophagic genes Atg5, Atg7 and BECN1 reversed Puerarin-inhibited OCP autophagy and proliferation. What’s more, RANKL could promote the autography of OCPs, which was recovered by Puerarin treatment. Interestingly, different from single-Puerarin treatment, we found that in the presence of RANKL, only BECN1 overexpression significantly reversed Puerarin-inhibited osteoclast differentiation and OCP autophagy. Conclusion In conclusion, Puerarin could inhibit the OCP autophagy in the presence or absence of RANKL, which blocked the OCP proliferation and osteoclast differentiation respectively. Moreover, BECN1 plays an essential role in Puerarin-inhibited osteoclastogenesis. Our study provides potential clue to further complete the intrinsic mechanism of Puerarin in treating osteoporosis.
Collapse
|