1
|
Xia Y, Wang B, Pan P, Ren X, Gao L, Xiong J, Ma Y. [Ferroptosis-related genes in osteoporosis: a bioinformatics analysis and in vitro study]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:680-690. [PMID: 39608795 DOI: 10.3724/zdxbyxb-2024-0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
OBJECTIVES To explore ferroptosis-related genes in osteoporosis through bioinformatic analysis and in vitro study. METHODS Osteoporosis-related genes were identified from dataset GSE35958 in the Gene Expression Omnibus database; and the ferroptosis-related genes were identified from the FerrDb database. These were intersected with the differentially expressed genes in GSE35958 to obtain ferroptosis-related genes in osteoporosis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed for the differentially expressed genes. And Spearman correlation and protein-protein interaction network analysis were performed. Then, the hub genes of ferroptosis in osteoporosis were screened by Degree, MNC, EPC, MCC and DMNC in Cytoscape software CytoHubba plugin; and analyzed with receiver operating characteristic (ROC) curves. The bone marrow mesenchymal stem cells from osteoporosis patients (osteoporosis group) and non-osteoporosis patients (control group) were subjected to quantitative reverse transcription polymerase chain reaction to detect the messenger RNA expression of ferroptosis hub genes in both groups. RESULTS A total of 32 differentially expressed genes related to ferroptosis in osteoporosis were identified, including 26 up-regulated genes and 6 down-regulated genes. GO enrichment analysis showed that the identified genes were mainly involved in intercellular adhesion, lipid metabolism and cytokine response. KEGG enrichment analysis showed that the genes were mainly involved in signaling pathways of adhesive plaques, MAPK, PI3K-Akt, and Wnt. Spearman correlation analysis showed correlation among differentially expressed genes. Six hub genes for ferroptosis in osteoporosis were obtained, namely MAPK3, CDKN1A, MAP1LC3A, TNF, RELA, and TGF-β1. ROC curve analysis showed that these hub genes had good diagnostic performance in osteoporosis and may become potential biomarkers of osteoporosis. In vitro experiments confirmed significant differences in these hub genes between the control group and the osteoporosis group (all P<0.05). CONCLUSIONS This study has identified six ferroptosis-related hub genes in osteoporosis, which may be used as novel biomarkers for the early diagnosis and treatment of osteoporosis.
Collapse
Affiliation(s)
- Yushuang Xia
- The First Clinical College of Hubei University of Chinese Medicine, Wuhan 430061, China.
| | - Bo Wang
- Rehabilitation Medicine Center, Wuhan First Municipal Hospital Affiliated to Hubei University of Chinese Medicine, Wuhan 430022, China
| | - Pengfei Pan
- The First Clinical College of Hubei University of Chinese Medicine, Wuhan 430061, China
| | - Xiangshun Ren
- Rehabilitation Medicine Center, Wuhan First Municipal Hospital Affiliated to Hubei University of Chinese Medicine, Wuhan 430022, China
| | - Lixi Gao
- Rehabilitation Medicine Center, Wuhan First Municipal Hospital Affiliated to Hubei University of Chinese Medicine, Wuhan 430022, China
| | - Jian Xiong
- Rehabilitation Medicine Center, Wuhan First Municipal Hospital Affiliated to Hubei University of Chinese Medicine, Wuhan 430022, China
| | - Yan Ma
- Rehabilitation Medicine Center, Wuhan First Municipal Hospital Affiliated to Hubei University of Chinese Medicine, Wuhan 430022, China.
| |
Collapse
|
2
|
Yin Y, Tang Q, Yang J, Gui S, Zhang Y, Shen Y, Zhou X, Yu S, Chen G, Sun J, Han Z, Zhang L, Chen L. Endothelial BMAL1 decline during aging leads to bone loss by destabilizing extracellular fibrillin-1. J Clin Invest 2024; 134:e176660. [PMID: 39680455 DOI: 10.1172/jci176660] [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: 10/13/2023] [Accepted: 10/15/2024] [Indexed: 12/18/2024] Open
Abstract
The occurrence of aging is intricately associated with alterations in circadian rhythms that coincide with stem cell exhaustion. Nonetheless, the extent to which the circadian system governs skeletal aging remains inadequately understood. Here, we noticed that skeletal aging in male mice was accompanied by a decline in a core circadian protein, BMAL1, especially in bone marrow endothelial cells (ECs). Using male mice with endothelial KO of aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1), we ascertained that endothelial BMAL1 in bone played a crucial role in ensuring the stability of an extracellular structural component, fibrillin-1 (FBN1), through regulation of the equilibrium between the extracellular matrix (ECM) proteases thrombospondin type 1 domain-containing protein 4 (THSD4) and metalloproteinase with thrombospondin motifs 4 (ADAMTS4), which promote FBN1 assembly and breakdown, respectively. The decline of endothelial BMAL1 during aging prompted excessive breakdown of FBN1, leading to persistent activation of TGF-β/SMAD3 signaling and exhaustion of bone marrow mesenchymal stem cells. Meanwhile, the free TGF-β could promote osteoclast formation. Further analysis revealed that activation of ADAMTS4 in ECs lacking BMAL1 was stimulated by TGF-β/SMAD3 signaling through an ECM-positive feedback mechanism, whereas THSD4 was under direct transcriptional control by endothelial BMAL1. Our investigation has elucidated the etiology of bone aging in male mice by defining the role of ECs in upholding the equilibrium within the ECM, consequently coordinating osteogenic and osteoclastic activities and retarding skeletal aging.
Collapse
Affiliation(s)
- Ying Yin
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Qingming Tang
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Jingxi Yang
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Shiqi Gui
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yifan Zhang
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yufeng Shen
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Xin Zhou
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Shaoling Yu
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Guangjin Chen
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Jiwei Sun
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Zhenshuo Han
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Chen
- Department of Stomatology, Union Hospital and
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| |
Collapse
|
3
|
Herro R, Grimes HL. The diverse roles of neutrophils from protection to pathogenesis. Nat Immunol 2024; 25:2209-2219. [PMID: 39567761 DOI: 10.1038/s41590-024-02006-5] [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: 05/30/2024] [Accepted: 10/07/2024] [Indexed: 11/22/2024]
Abstract
Neutrophil granulocytes are the most abundant leukocytes in the blood and constitute a critical arm of innate immunity. They are generated in the bone marrow, and under homeostatic conditions enter the bloodstream to patrol tissues and scout for potential pathogens that they quickly destroy through phagocytosis, intracellular degradation, release of granules and formation of extracellular traps. Thus, neutrophils are important effector cells involved in antibacterial defense. However, neutrophils can also be pathogenic. Emerging data suggest they have critical functions related to tissue repair and fibrosis. Moreover, similarly to other innate immune cells, neutrophil cell states are affected by their microenvironment. Notably, this includes tumors that co-opt neutrophils. Neutrophils can undergo transcriptional and epigenetic reprogramming, thus causing or modulating inflammation and injury. It is also possible that distinct neutrophil subsets are generated with designated functions in the bone marrow. Understanding neutrophil plasticity and alternative cell states will help resolve their contradictive roles. This Review summarizes the most recent key findings surrounding protective versus pathogenic functions of neutrophils; elaborating on phenotype-specific subsets of neutrophils and their involvement in homeostasis and disease.
Collapse
Affiliation(s)
- Rana Herro
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA.
| | - H Leighton Grimes
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
4
|
Chermside-Scabbo CJ, Shuster JT, Erdmann-Gilmore P, Tycksen E, Zhang Q, Townsend RR, Silva MJ. A proteomics approach to study mouse long bones: examining baseline differences and mechanical loading-induced bone formation in young-adult and old mice. Aging (Albany NY) 2024; 16:12726-12768. [PMID: 39400554 PMCID: PMC11501390 DOI: 10.18632/aging.206131] [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: 06/20/2023] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
With aging, bone mass declines and the anabolic effects of skeletal loading diminish. While much research has focused on gene transcription, how bone ages and loses its mechanoresponsiveness at the protein level remains unclear. We developed a novel proteomics approach and performed a paired mass spectrometry and RNA-seq analysis on tibias from young-adult (5-month) and old (22-month) mice. We report the first correlation estimate between the bone proteome and transcriptome (Spearman ρ = 0.40), which is in line with other tissues but indicates that a relatively low amount of variation in protein levels is explained by the variation in transcript levels. Of 71 shared targets that differed with age, eight were associated with bone mineral density in previous GWAS, including understudied targets Asrgl1 and Timp2. We used complementary RNA in situ hybridization to confirm that Asrgl1 and Timp2 had reduced expression in osteoblasts/osteocytes in old bones. We also found evidence for reduced TGF-beta signaling with aging, in particular Tgfb2. Next, we defined proteomic changes following mechanical loading. At the protein level, bone differed more with age than with loading, and aged bone had fewer loading-induced changes. Overall, our findings underscore the need for complementary protein-level assays in skeletal biology research.
Collapse
Affiliation(s)
- Christopher J. Chermside-Scabbo
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John T. Shuster
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Petra Erdmann-Gilmore
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric Tycksen
- Department of Genetics, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Qiang Zhang
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - R. Reid Townsend
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| |
Collapse
|
5
|
Yao Z, Ayoub A, Srinivasan V, Wu J, Tang C, Duan R, Milosavljevic A, Xing L, Ebetino FH, Frontier AJ, Boyce BF. Hydroxychloroquine and a low antiresorptive activity bisphosphonate conjugate prevent and reverse ovariectomy-induced bone loss in mice through dual antiresorptive and anabolic effects. Bone Res 2024; 12:52. [PMID: 39231935 PMCID: PMC11375055 DOI: 10.1038/s41413-024-00352-6] [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: 04/20/2024] [Revised: 06/14/2024] [Accepted: 07/12/2024] [Indexed: 09/06/2024] Open
Abstract
Osteoporosis remains incurable. The most widely used antiresorptive agents, bisphosphonates (BPs), also inhibit bone formation, while the anabolic agent, teriparatide, does not inhibit bone resorption, and thus they have limited efficacy in preventing osteoporotic fractures and cause some side effects. Thus, there is an unmet need to develop dual antiresorptive and anabolic agents to prevent and treat osteoporosis. Hydroxychloroquine (HCQ), which is used to treat rheumatoid arthritis, prevents the lysosomal degradation of TNF receptor-associated factor 3 (TRAF3), an NF-κB adaptor protein that limits bone resorption and maintains bone formation. We attempted to covalently link HCQ to a hydroxyalklyl BP (HABP) with anticipated low antiresorptive activity, to target delivery of HCQ to bone to test if this targeting increases its efficacy to prevent TRAF3 degradation in the bone microenvironment and thus reduce bone resorption and increase bone formation, while reducing its systemic side effects. Unexpectedly, HABP-HCQ was found to exist as a salt in aqueous solution, composed of a protonated HCQ cation and a deprotonated HABP anion. Nevertheless, it inhibited osteoclastogenesis, stimulated osteoblast differentiation, and increased TRAF3 protein levels in vitro. HABP-HCQ significantly inhibited both osteoclast formation and bone marrow fibrosis in mice given multiple daily PTH injections. In contrast, HCQ inhibited marrow fibrosis, but not osteoclast formation, while the HABP alone inhibited osteoclast formation, but not fibrosis, in the mice. HABP-HCQ, but not HCQ, prevented trabecular bone loss following ovariectomy in mice and, importantly, increased bone volume in ovariectomized mice with established bone loss because HABP-HCQ increased bone formation and decreased bone resorption parameters simultaneously. In contrast, HCQ increased bone formation, but did not decrease bone resorption parameters, while HABP also restored the bone lost in ovariectomized mice, but it inhibited parameters of both bone resorption and formation. Our findings suggest that the combination of HABP and HCQ could have dual antiresorptive and anabolic effects to prevent and treat osteoporosis.
Collapse
Affiliation(s)
- Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| | - Akram Ayoub
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | | | - Jun Wu
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Churou Tang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- School of Arts and Sciences, University of Rochester, Rochester, NY14627, USA
| | - Rong Duan
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | | | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Frank H Ebetino
- Department of Chemistry, University of Rochester, Rochester, NY14627, USA
- BioVinc, LLC, Pasadena, CA, 91107, USA
| | - Alison J Frontier
- Department of Chemistry, University of Rochester, Rochester, NY14627, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
6
|
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
|
7
|
Wu Z, Li W, Jiang K, Lin Z, Qian C, Wu M, Xia Y, Li N, Zhang H, Xiao H, Bai J, Geng D. Regulation of bone homeostasis: signaling pathways and therapeutic targets. MedComm (Beijing) 2024; 5:e657. [PMID: 39049966 PMCID: PMC11266958 DOI: 10.1002/mco2.657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
As a highly dynamic tissue, bone is continuously rebuilt throughout life. Both bone formation by osteoblasts and bone resorption by osteoclasts constitute bone reconstruction homeostasis. The equilibrium of bone homeostasis is governed by many complicated signaling pathways that weave together to form an intricate network. These pathways coordinate the meticulous processes of bone formation and resorption, ensuring the structural integrity and dynamic vitality of the skeletal system. Dysregulation of the bone homeostatic regulatory signaling network contributes to the development and progression of many skeletal diseases. Significantly, imbalanced bone homeostasis further disrupts the signaling network and triggers a cascade reaction that exacerbates disease progression and engenders a deleterious cycle. Here, we summarize the influence of signaling pathways on bone homeostasis, elucidating the interplay and crosstalk among them. Additionally, we review the mechanisms underpinning bone homeostatic imbalances across diverse disease landscapes, highlighting current and prospective therapeutic targets and clinical drugs. We hope that this review will contribute to a holistic understanding of the signaling pathways and molecular mechanisms sustaining bone homeostasis, which are promising to contribute to further research on bone homeostasis and shed light on the development of targeted drugs.
Collapse
Affiliation(s)
- Zebin Wu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Wenming Li
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Kunlong Jiang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Zhixiang Lin
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Chen Qian
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Mingzhou Wu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Yu Xia
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Ning Li
- Department of OrthopedicsCentre for Leading Medicine and Advanced Technologies of IHMDivision of Life Sciences and MedicineThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Hongtao Zhang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Haixiang Xiao
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Department of OrthopedicsJingjiang People's HospitalSeventh Clinical Medical School of Yangzhou UniversityJingjiangJiangsu ProvinceChina
| | - Jiaxiang Bai
- Department of OrthopedicsCentre for Leading Medicine and Advanced Technologies of IHMDivision of Life Sciences and MedicineThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Dechun Geng
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
8
|
Wei E, Hu M, Wu L, Pan X, Zhu Q, Liu H, Liu Y. TGF-β signaling regulates differentiation of MSCs in bone metabolism: disputes among viewpoints. Stem Cell Res Ther 2024; 15:156. [PMID: 38816830 PMCID: PMC11140988 DOI: 10.1186/s13287-024-03761-w] [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: 11/07/2023] [Accepted: 05/14/2024] [Indexed: 06/01/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into cells of different lineages to form mesenchymal tissues, which are promising in regard to treatment for bone diseases. Their osteogenic differentiation is under the tight regulation of intrinsic and extrinsic factors. Transforming growth factor β (TGF-β) is an essential growth factor in bone metabolism, which regulates the differentiation of MSCs. However, published studies differ in their views on whether TGF-β signaling regulates the osteogenic differentiation of MSCs positively or negatively. The controversial results have not been summarized systematically and the related explanations are required. Therefore, we reviewed the basics of TGF-β signaling and summarized how each of three isoforms regulates osteogenic differentiation. Three isoforms of TGF-β (TGF-β1/β2/β3) play distinct roles in regulating osteogenic differentiation of MSCs. Additionally, other possible sources of conflicts are summarized here. Further understanding of TGF-β signaling regulation in MSCs may lead to new applications to promote bone regeneration and improve therapies for bone diseases.
Collapse
Affiliation(s)
- Erfan Wei
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Central Laboratory, Peking University School and Hospital of Stomatology , No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Menglong Hu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Central Laboratory, Peking University School and Hospital of Stomatology , No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Likun Wu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Central Laboratory, Peking University School and Hospital of Stomatology , No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Xingtong Pan
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Central Laboratory, Peking University School and Hospital of Stomatology , No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Qiyue Zhu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Central Laboratory, Peking University School and Hospital of Stomatology , No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Hao Liu
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials , Peking University School and Hospital of Stomatology, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Central Laboratory, Peking University School and Hospital of Stomatology , No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
| |
Collapse
|
9
|
Yao Z, Ayoub A, Srinivasan V, Wu J, Tang C, Duan R, Milosavljevic A, Ebetino F, Frontier A, Boyce B. Hydroxychloroquine and a low activity bisphosphonate conjugate prevent and reverse ovariectomy-induced bone loss in mice through dual antiresorptive and anabolic effects. RESEARCH SQUARE 2024:rs.3.rs-4237258. [PMID: 38746138 PMCID: PMC11092802 DOI: 10.21203/rs.3.rs-4237258/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Osteoporosis is incurable because there are no dual antiresorptive and anabolic therapeutic agents that can be administered long-term. The most widely used antiresorptive agents, bisphosphonates (BPs), also inhibit bone formation and thus have limited effect in preventing osteoporotic fracture. Hydroxychloroquine (HCQ), which is used to treat rheumatoid arthritis, prevents the lysosomal degradation of TNF receptor-associated factor 3 (TRAF3), an NF-κB adaptor protein that limits bone resorption and maintains bone formation. We attempted to covalently link HCQ to a hydroxyalklyl BP (HABP) with anticipated low antiresorptive activity, to target delivery of HCQ to bone to test if this targeting increases its efficacy to prevent TRAF3 degradation in the bone microenvironment and thus reduce bone resorption and increase bone formation, while reducing its systemic side effects. Unexpectedly, HABP-HCQ was found to exist as a salt in aqueous solution, composed of a protonated HCQ cation and a deprotonated HABP anion. Nevertheless, it inhibited osteoclastogenesis, stimulated osteoblast differentiation, and increased TRAF3 protein levels in vitro. HABP-HCQ significantly inhibited both osteoclast formation and bone marrow fibrosis in mice given multiple daily PTH injections. In contrast, HCQ inhibited fibrosis, but not osteoclast formation, while the HABP alone inhibited osteoclast formation, but not fibrosis, in the mice. HABP-HCQ, but not HCQ, prevented trabecular bone loss following ovariectomy in mice and, importantly, increased bone volume in ovariectomized mice with established bone loss because HABP-HCQ increased bone formation and decreased bone resorption parameters simultaneously. In contrast, HCQ increased bone formation, but did not decrease bone resorption parameters, while HABP also restored the bone lost in ovariectomized mice, but it inhibited parameters of both bone resorption and formation. Our findings suggest that the combination of HABP and HCQ could have dual antiresorptive and anabolic effects to prevent and treat osteoporosis.
Collapse
Affiliation(s)
| | | | | | - Jun Wu
- University of Rochester Medical Center
| | | | | | | | | | | | | |
Collapse
|
10
|
Gao Y, Zou Y, Sokolowskei D, Xing X, Tower RJ, Lai Z, Shi J, Zhu L, Zheng Q, James AW, Xu J, Zhang Z. Nr4a1 enhances Wnt4 transcription to promote mesenchymal stem cell osteogenesis and alleviates inflammation-inhibited bone regeneration. Mol Ther 2024; 32:1479-1496. [PMID: 38429926 PMCID: PMC11081873 DOI: 10.1016/j.ymthe.2024.02.034] [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: 09/27/2023] [Revised: 01/15/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
Intense inflammatory response impairs bone marrow mesenchymal stem cell (BMSC)-mediated bone regeneration, with transforming growth factor (TGF)-β1 being the most highly expressed cytokine. However, how to find effective and safe means to improve bone formation impaired by excessive TGF-β1 remains unclear. In this study, we found that the expression of orphan nuclear receptor Nr4a1, an endogenous repressor of TGF-β1, was suppressed directly by TGF-β1-induced Smad3 and indirectly by Hdac4, respectively. Importantly, Nr4a1 overexpression promoted BMSC osteogenesis and reversed TGF-β1-mediated osteogenic inhibition and pro-fibrotic effects. Transcriptomic and histologic analyses confirmed that upregulation of Nr4a1 increased the transcription of Wnt family member 4 (Wnt4) and activated Wnt pathway. Mechanistically, Nr4a1 bound to the promoter of Wnt4 and regulated its expression, thereby enhancing the osteogenic capacity of BMSCs. Moreover, treatment with Nr4a1 gene therapy or Nr4a1 agonist Csn-B could promote ectopic bone formation, defect repair, and fracture healing. Finally, we demonstrated the correlation of NR4A1 with osteogenesis and the activation of the WNT4/β-catenin pathway in human BMSCs and fracture samples. Taken together, these findings uncover the critical role of Nr4a1 in bone formation and alleviation of inflammation-induced bone regeneration disorders, and suggest that Nr4a1 has the potential to be a therapeutic target for accelerating bone healing.
Collapse
Affiliation(s)
- Yangshuai Gao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yuming Zou
- Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China; Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Dimitri Sokolowskei
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Robert J Tower
- Center for Organogenesis, Regeneration and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zejia Lai
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiawei Shi
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Liheng Zhu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Quan Zheng
- Department of Orthopedic Surgery, Luan Hospital Affiliated to Anhui Medical University, Luan, Anhui 237001, China
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Jiajia Xu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China.
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
11
|
Ye J, Hua Z, Xiao J, Shao Y, Li S, Yin H, Wu M, Rong Y, Hong B, Guo Y, Ma Y, Wang J. p-Smad3 differentially regulates the cytological behavior of osteoclasts before and after osteoblasts maturation. Mol Biol Rep 2024; 51:525. [PMID: 38632128 DOI: 10.1007/s11033-024-09400-0] [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: 06/13/2023] [Accepted: 02/28/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND A series of previous investigations have revealed that p-Smad3 plays a facilitative role in the differentiation and maturation of osteoblasts, while also regulating the expression of certain intercellular communication factors. However, the effects of p-Smad3 in osteoblasts before and after maturation on the proliferation, migration, differentiation, apoptosis and other cellular behaviors of osteoclasts have not been reported. METHODS MC3T3-E1 cells were cultured in osteogenic induction medium for varying durations, After that, the corresponding conditioned medium was collected and the osteoclast lineage cells were treated. To elucidate the regulatory role of p-Smad3 within osteoblasts, we applied the activator TGF-β1 and inhibitor SIS3 to immature and mature osteoblasts and collected corresponding conditioned media for osteoclast intervention. RESULTS We observed an elevation of p-Smad3 and Smad3 during the early stage of osteoblast differentiation, followed by a decline in the later stage. we discovered that as osteoblasts mature, their conditioned media inhibit osteoclasts differentiation and the osteoclast-coupled osteogenic effect. However, it promotes apoptosis in osteoclasts and the angiogenesis coupled with osteoclasts. p-Smad3 in immature osteoblasts, through paracrine effects, promotes the migration, differentiation, and osteoclast-coupled osteogenic effects of osteoclast lineage cells. For mature osteoblasts, p-Smad3 facilitates osteoclast apoptosis and the angiogenesis coupled with osteoclasts. CONCLUSIONS As pre-osteoblasts undergo maturation, p-Smad3 mediated a paracrine effect that transitions osteoclast cellular behaviors from inducing differentiation and stimulating bone formation to promoting apoptosis and coupling angiogenesis.
Collapse
Affiliation(s)
- Jiapeng Ye
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Zhen Hua
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Jirimutu Xiao
- Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
- Mongolian Medicine College, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010159, China
| | - Yang Shao
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Shaoshuo Li
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Heng Yin
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Mao Wu
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Yi Rong
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Bowen Hong
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Yang Guo
- Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Yong Ma
- Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Jianwei Wang
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China.
| |
Collapse
|
12
|
Wang Y, Yang C, Hou Y, Wang J, Zhang K, Wang L, Sun D, Li X, Wei R, Nian H. Dimethyl itaconate inhibits antigen-specific Th17 cell responses and autoimmune inflammation via modulating NRF2/STAT3 signaling. FASEB J 2024; 38:e23607. [PMID: 38581245 DOI: 10.1096/fj.202302293rr] [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: 11/09/2023] [Revised: 03/14/2024] [Accepted: 03/29/2024] [Indexed: 04/08/2024]
Abstract
Pathogenic Th17 cells play a crucial role in autoimmune diseases like uveitis and its animal model, experimental autoimmune uveitis (EAU). Dimethyl itaconate (DMI) possesses potent anti-inflammatory effects. However, there is still a lack of knowledge about the role of DMI in regulating pathogenic Th17 cells and EAU. Here, we reported that intraperitoneal administration of DMI significantly inhibited the severity of EAU via selectively suppressing Th17 cell responses. In vitro antigen stimulation studies revealed that DMI dramatically decreased the frequencies and function of antigen-specific Th17, but not Th1, cells. Moreover, DMI hampered the differentiation of naive CD4+ T cells toward pathogenic Th17 cells. DMI-treated DCs produced less IL-1β, IL-6, and IL-23, and displayed an impaired ability to stimulate antigen-specific Th17 activation. Mechanistically, DMI activated the NRF2/HO-1 pathway and suppressed STAT3 signaling, which subsequently restrains p-STAT3 nuclear translocation, leading to decreased pathogenic Th17 cell responses. Thus, we have identified an important role for DMI in regulating pathogenic Th17 cells, supporting DMI as a promising therapy in Th17 cell-driven autoimmune diseases including uveitis.
Collapse
Affiliation(s)
- Ying Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Chao Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Yubiao Hou
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Jiali Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Kailang Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Lihua Wang
- Department of Kidney Diseases and Blood Purification, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Deming Sun
- Doheny Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Ruihua Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Hong Nian
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| |
Collapse
|
13
|
Zhao Z, Du Y, Yan K, Zhang L, Guo Q. Exercise and osteoimmunology in bone remodeling. FASEB J 2024; 38:e23554. [PMID: 38588175 DOI: 10.1096/fj.202301508rrr] [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: 07/25/2023] [Revised: 02/20/2024] [Accepted: 02/28/2024] [Indexed: 04/10/2024]
Abstract
Bones can form the scaffolding of the body, support the organism, coordinate somatic movements, and control mineral homeostasis and hematopoiesis. The immune system plays immune supervisory, defensive, and regulatory roles in the organism, which mainly consists of immune organs (spleen, bone marrow, tonsils, lymph nodes, etc.), immune cells (granulocytes, platelets, lymphocytes, etc.), and immune molecules (immune factors, interferons, interleukins, tumor necrosis factors, etc.). Bone and the immune system have long been considered two distinct fields of study, and the bone marrow, as a shared microenvironment between the bone and the immune system, closely links the two. Osteoimmunology organically combines bone and the immune system, elucidates the role of the immune system in bone, and creatively emphasizes its interdisciplinary characteristics and the function of immune cells and factors in maintaining bone homeostasis, providing new perspectives for skeletal-related field research. In recent years, bone immunology has gradually become a hot spot in the study of bone-related diseases. As a new branch of immunology, bone immunology emphasizes that the immune system can directly or indirectly affect bones through the RANKL/RANK/OPG signaling pathway, IL family, TNF-α, TGF-β, and IFN-γ. These effects are of great significance for understanding inflammatory bone loss caused by various autoimmune or infectious diseases. In addition, as an external environment that plays an important role in immunity and bone, this study pays attention to the role of exercise-mediated bone immunity in bone reconstruction.
Collapse
Affiliation(s)
- Zhonghan Zhao
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Yuxiang Du
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Kai Yan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Lingli Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai, China
| | - Qiang Guo
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
14
|
Zhao S, Qiao Z, Pfeifer R, Pape HC, Mao K, Tang H, Meng B, Chen S, Liu H. Modulation of fracture healing by senescence-associated secretory phenotype (SASP): a narrative review of the current literature. Eur J Med Res 2024; 29:38. [PMID: 38195489 PMCID: PMC10775505 DOI: 10.1186/s40001-023-01604-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 12/19/2023] [Indexed: 01/11/2024] Open
Abstract
The senescence-associated secretory phenotype (SASP) is a generic term for the secretion of cytokines, such as pro-inflammatory factors and proteases. It is a crucial feature of senescent cells. SASP factors induce tissue remodeling and immune cell recruitment. Previous studies have focused on the beneficial role of SASP during embryonic development, wound healing, tissue healing in general, immunoregulation properties, and cancer. However, some recent studies have identified several negative effects of SASP on fracture healing. Senolytics is a drug that selectively eliminates senescent cells. Senolytics can inhibit the function of senescent cells and SASP, which has been found to have positive effects on a variety of aging-related diseases. At the same time, recent data suggest that removing senescent cells may promote fracture healing. Here, we reviewed the latest research progress about SASP and illustrated the inflammatory response and the influence of SASP on fracture healing. This review aims to understand the role of SASP in fracture healing, aiming to provide an important clinical prevention and treatment strategy for fracture. Clinical trials of some senolytics agents are underway and are expected to clarify the effectiveness of their targeted therapy in the clinic in the future. Meanwhile, the adverse effects of this treatment method still need further study.
Collapse
Affiliation(s)
- Shangkun Zhao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhi Qiao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Roman Pfeifer
- Department of Traumatology, University Hospital of Zurich, Zurich, 8091, China
| | - Hans-Christoph Pape
- Department of Traumatology, University Hospital of Zurich, Zurich, 8091, China
| | - Keya Mao
- Chinese PLA General Hospital Beijing, Beijing, 100853, China
| | - Hai Tang
- Beijing Friendship Hospital, Beijing, 100050, China
| | - Bin Meng
- First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Songfeng Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongjian Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
15
|
Alake SE, Ice J, Robinson K, Price P, Hatter B, Wozniak K, Lin D, Chowanadisai W, Smith BJ, Lucas EA. Reduced estrogen signaling contributes to bone loss and cardiac dysfunction in interleukin-10 knockout mice. Physiol Rep 2024; 12:e15914. [PMID: 38217044 PMCID: PMC10787104 DOI: 10.14814/phy2.15914] [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: 08/28/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 01/14/2024] Open
Abstract
Characterization of the interleukin (IL)-10 knockout (KO) mouse with chronic gut inflammation, cardiovascular dysfunction, and bone loss suggests a critical role for this cytokine in interorgan communication within the gut, bone, and cardiovascular axis. We sought to understand the role of IL-10 in the cross-talk between these systems. Six-week-old IL-10 KO mice and their wild type (WT) counterparts were maintained on a standard rodent diet for 3 or 6 months. Gene expression of proinflammatory markers and Fgf23, serum 17β-estradiol (E2), and cardiac protein expression were assessed. Ileal Il17a and Tnf mRNA increased while Il6 mRNA increased in the bone and heart by at least 2-fold in IL-10 KO mice. Bone Dmp1 and Phex mRNA were repressed at 6 months in IL-10 KO mice, resulting in increased Fgf23 mRNA (~4-fold) that contributed to increased fibrosis. In the IL-10 KO mice, gut bacterial β-glucuronidase activity and ovarian Cyp19a1 mRNA were lower (p < 0.05), consistent with reduced serum E2 and reduced cardiac pNOS3 (Ser1119 ) in these mice. Treatment of ileal lymphocytes with E2 reduced gut inflammation in WT but not IL-10 KO mice. In conclusion, our data suggest that diminished estrogen and defective bone mineralization increased FGF23 which contributed to cardiac fibrosis in the IL-10 KO mouse.
Collapse
Affiliation(s)
- Sanmi E. Alake
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - John Ice
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Kara Robinson
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Payton Price
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Bethany Hatter
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Karen Wozniak
- Department of Microbiology and Molecular GeneticsOklahoma State UniversityStillwaterOklahomaUSA
| | - Dingbo Lin
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Winyoo Chowanadisai
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Brenda J. Smith
- Department of Obstetrics and GynecologyIndiana School of MedicineIndianapolisIndianaUSA
- Indiana Center for Musculoskeletal HealthIndiana School of MedicineIndianapolisIndianaUSA
| | - Edralin A. Lucas
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| |
Collapse
|
16
|
Cai M, Peng H, Liu M, Huang M, Zheng W, Zhang G, Lai W, Liao C, Cai L, Zhang D, Liu X. Vascular Pericyte-Derived Exosomes Inhibit Bone Resorption via Traf3. Int J Nanomedicine 2023; 18:7065-7077. [PMID: 38046234 PMCID: PMC10693246 DOI: 10.2147/ijn.s438229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/17/2023] [Indexed: 12/05/2023] Open
Abstract
Purpose Blood vessels distribute cells, oxygen, and nutrients throughout the body to support tissue growth and balance. Pericytes and endothelial cells form the inner wall of blood vessels, crucial for organ development and tissue homeostasis by producing paracrine signaling molecules. In the skeletal system, pericyte-derived vascular factors along with angiogenic factors released by bone cells regulate angiogenesis and bone formation. Although the involvement of angiogenic factors and skeletal blood vessels in bone homeostasis is relatively clear, the role of pericytes and the underlying mechanisms remain unknown. Here, our objective was to elucidate the significance of pericytes in regulating osteoclast differentiation. Methods We used tissue staining to detect the coverage of pericytes and osteoclasts in femoral tissues of osteoporotic mice and mice of different ages, analyzing their correlation. We developed mice with conditionally deleted pericytes, observing changes in bone mass and osteoclast activity using micro-computer tomography and tissue staining to detect the regulatory effect of pericytes on osteoclasts. Pericytes-derived exosomes (PC-EVs) were collected and co-cultured with monocytes that induce osteoclast differentiation to detect the effect of the former on the exosomes. Finally, the specific mechanism of PC-EVs regulating osteoclast differentiation was verified using RNA sequencing and Western blotting. Results Our study indicates a significant correlation between pericytes and age-related bone resorption. Conditional deletion of pericytes activated bone resorption and led to osteopenia in vivo. We discovered that PC-EVs inhibited the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway, which is mediated by tumor necrosis factor receptor-associated factor 3 (Traf3), negatively regulating osteoclast development and bone resorption. Silencing Traf3 in PC-EVs canceled their inhibitory effect on osteoclast differentiation. Conclusion Our study provides a novel perspective into the regulatory role of pericytes on bone resorption and may provide potential strategies for developing novel anti-bone resorption therapies.
Collapse
Affiliation(s)
- Mingxiang Cai
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Huizhen Peng
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Minyi Liu
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Maohua Huang
- College of Pharmacy, Jinan University, Guangzhou, 510632, People’s Republic of China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Wen Zheng
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Guilan Zhang
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Wenjia Lai
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Chufang Liao
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Lizhao Cai
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Dongmei Zhang
- College of Pharmacy, Jinan University, Guangzhou, 510632, People’s Republic of China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Xiangning Liu
- The First Affiliated Hospital of Jinan University, School of Stomatology, Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, 510630, People’s Republic of China
| |
Collapse
|
17
|
Boyce BF, Li J, Yao Z, Xing L. Nuclear Factor-Kappa B Regulation of Osteoclastogenesis and Osteoblastogenesis. Endocrinol Metab (Seoul) 2023; 38:504-521. [PMID: 37749800 PMCID: PMC10613774 DOI: 10.3803/enm.2023.501] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 09/27/2023] Open
Abstract
Maintenance of skeletal integrity requires the coordinated activity of multinucleated bone-resorbing osteoclasts and bone-forming osteoblasts. Osteoclasts form resorption lacunae on bone surfaces in response to cytokines by fusion of precursor cells. Osteoblasts are derived from mesenchymal precursors and lay down new bone in resorption lacunae during bone remodeling. Nuclear factorkappa B (NF-κB) signaling regulates osteoclast and osteoblast formation and is activated in osteoclast precursors in response to the essential osteoclastogenic cytokine, receptor activator of NF-κB ligand (RANKL), which can also control osteoblast formation through RANK-RANKL reverse signaling in osteoblast precursors. RANKL and some pro-inflammatory cytokines, including tumor necrosis factor (TNF), activate NF-κB signaling to positively regulate osteoclast formation and functions. However, these cytokines also limit osteoclast and osteoblast formation through NF-κB signaling molecules, including TNF receptor-associated factors (TRAFs). TRAF6 mediates RANKL-induced osteoclast formation through canonical NF-κB signaling. In contrast, TRAF3 limits RANKL- and TNF-induced osteoclast formation, and it restricts transforming growth factor β (TGFβ)-induced inhibition of osteoblast formation in young and adult mice. During aging, neutrophils expressing TGFβ and C-C chemokine receptor type 5 (CCR5) increase in bone marrow of mice in response to increased NF-κB-induced CC motif chemokine ligand 5 (CCL5) expression by mesenchymal progenitor cells and injection of these neutrophils into young mice decreased bone mass. TGFβ causes degradation of TRAF3, resulting in decreased glycogen synthase kinase-3β/β-catenin-mediated osteoblast formation and age-related osteoporosis in mice. The CCR5 inhibitor, maraviroc, prevented accumulation of TGFβ+/CCR5+ neutrophils in bone marrow and increased bone mass by inhibiting bone resorption and increasing bone formation in aged mice. This paper updates current understanding of how NF-κB signaling is involved in the positive and negative regulation of cytokine-mediated osteoclast and osteoblast formation and activation with a focus on the role of TRAF3 signaling, which can be targeted therapeutically to enhance bone mass.
Collapse
Affiliation(s)
- Brendan F. Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Jinbo Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
18
|
Yu R, Han H, Chu S, Ding Y, Jin S, Wang Y, Jiang W, Liu Y, Zou Y, Wang M, Liu Q, Sun G, Jiang B, Gong Y. CUL4B orchestrates mesenchymal stem cell commitment by epigenetically repressing KLF4 and C/EBPδ. Bone Res 2023; 11:29. [PMID: 37268647 DOI: 10.1038/s41413-023-00263-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 06/04/2023] Open
Abstract
Dysregulated lineage commitment of mesenchymal stem cells (MSCs) contributes to impaired bone formation and an imbalance between adipogenesis and osteogenesis during skeletal aging and osteoporosis. The intrinsic cellular mechanism that regulates MSC commitment remains unclear. Here, we identified Cullin 4B (CUL4B) as a critical regulator of MSC commitment. CUL4B is expressed in bone marrow MSCs (BMSCs) and downregulated with aging in mice and humans. Conditional knockout of Cul4b in MSCs resulted in impaired postnatal skeletal development with low bone mass and reduced bone formation. Moreover, depletion of CUL4B in MSCs aggravated bone loss and marrow adipose accumulation during natural aging or after ovariectomy. In addition, CUL4B deficiency in MSCs reduced bone strength. Mechanistically, CUL4B promoted osteogenesis and inhibited adipogenesis of MSCs by repressing KLF4 and C/EBPδ expression, respectively. The CUL4B complex directly bound to Klf4 and Cebpd and epigenetically repressed their transcription. Collectively, this study reveals CUL4B-mediated epigenetic regulation of the osteogenic or adipogenic commitment of MSCs, which has therapeutic implications in osteoporosis.
Collapse
Affiliation(s)
- Ruiqi Yu
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Hong Han
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shuxian Chu
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yijun Ding
- The Key Laboratory of Liquid‒Solid Structural Evolution and Processing of Materials of Ministry of Education and Institute of Liquid Metal and Casting Technology, School of Materials Science and Engineering, Shandong University, Jinan, 250012, China
| | - Shiqi Jin
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yufeng Wang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Wei Jiang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yuting Liu
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Molin Wang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qiao Liu
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Gongping Sun
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
19
|
The Roles of TRAF3 in Immune Responses. DISEASE MARKERS 2023; 2023:7787803. [PMID: 36845015 PMCID: PMC9949957 DOI: 10.1155/2023/7787803] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/18/2023]
Abstract
Seven tumor necrosis factor receptor- (TNFR-) associated factors (TRAFs) have been found in mammals, which are primarily involved in the signal translation of the TNFR superfamily, the Toll-like receptor (TLR) family, and the retinoic acid-inducible gene I- (RIG-I-) like receptor (RLR) family. TRAF3 is one of the most diverse members of the TRAF family. It can positively regulate type I interferon production while negatively regulating signaling pathways of classical nuclear factor-κB, nonclassical nuclear factor-κB, and mitogen-activated protein kinase (MAPK). This review summarizes the roles of TRAF3 signaling and the related immune receptors (e.g., TLRs) in several preclinical and clinical diseases and focuses on the roles of TRAF3 in immune responses, the regulatory mechanisms, and its role in disease.
Collapse
|
20
|
Liu J, Lin X, McDavid A, Yang Y, Zhang H, Boyce BF, Xing L. Molecular signatures distinguish senescent cells from inflammatory cells in aged mouse callus stromal cells. Front Endocrinol (Lausanne) 2023; 14:1090049. [PMID: 36875448 PMCID: PMC9981154 DOI: 10.3389/fendo.2023.1090049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/02/2023] [Indexed: 02/18/2023] Open
Abstract
Cellular senescence plays important roles in age-related diseases, including musculoskeletal disorders. Senescent cells (SCs) exert a senescence-associated secretory phenotype (SASP) by producing SASP factors, some of which overlap with factors produced by inflammatory cells (Inf-Cs). However, the differences between SCs and Inf-Cs and how they interact with each other during fracture repair have not been well studied. Here, we analyzed single cell RNA sequencing data of aged mouse fracture callus stromal cells. We defined Inf-Cs as cells that express NF-κB Rela/Relb, SCs as cells that express the senescence genes, Cdkn1a, Cdkn2a or Cdkn2c, and inflammatory SCs (Inf-SCs) as cells that express both NF-κB and senescence genes. Differentially expressed genes and pathway analyses revealed that Inf-SCs and SCs had a similar gene expression profile and upregulated pathways that are related to DNA damage/oxidation-reduction and cellular senescence, while Inf-Cs expressed different gene signatures and pathways from SCs and Inf-SCs, mainly related to inflammation. Cellchat software analysis indicated that SCs and Inf-SCs are potential ligand-producing cells that affect Inf-Cs as target cells. Cell culture experiments demonstrated that SC conditioned medium promoted inflammatory gene expression by callus-derived mesenchymal progenitor cells, and Inf-Cs had reduced osteoblast differentiation capacity. In summary, we have identified three cell subclusters associated with inflammation and senescence in callus stromal cells, predicted potential effects of Inf-SCs and SCs on Inf-Cs by production of active ligands, and demonstrated that when mesenchymal progenitors acquire inflammatory phenotypes their osteogenic potential is reduced.
Collapse
Affiliation(s)
- Jiatong Liu
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Xi Lin
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Andrew McDavid
- Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, United States
| | - Yutiancheng Yang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Hengwei Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Brendan F. Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
- Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
- Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
21
|
Li J, Yao Z, Liu X, Duan R, Yi X, Ayoub A, Sanders JO, Mesfin A, Xing L, Boyce BF. TGFβ1 +CCR5 + neutrophil subset increases in bone marrow and causes age-related osteoporosis in male mice. Nat Commun 2023; 14:159. [PMID: 36631487 PMCID: PMC9834218 DOI: 10.1038/s41467-023-35801-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
TGFβ1 induces age-related bone loss by promoting degradation of TNF receptor-associated factor 3 (TRAF3), levels of which decrease in murine and human bone during aging. We report that a subset of neutrophils (TGFβ1+CCR5+) is the major source of TGFβ1 in murine bone. Their numbers are increased in bone marrow (BM) of aged wild-type mice and adult mice with TRAF3 conditionally deleted in mesenchymal progenitor cells (MPCs), associated with increased expression in BM of the chemokine, CCL5, suggesting that TRAF3 in MPCs limits TGFβ1+CCR5+ neutrophil numbers in BM of young mice. During aging, TGFβ1-induced TRAF3 degradation in MPCs promotes NF-κB-mediated expression of CCL5 by MPCs, associated with higher TGFβ1+CCR5+ neutrophil numbers in BM where they induce bone loss. TGFβ1+CCR5+ neutrophils decreased bone mass in male mice. The FDA-approved CCR5 antagonist, maraviroc, reduced TGFβ1+CCR5+ neutrophil numbers in BM and increased bone mass in aged mice. 15-mon-old mice with TGFβRII specifically deleted in MPCs had lower numbers of TGFβ1+CCR5+ neutrophils in BM and higher bone volume than wild-type littermates. We propose that pharmacologic reduction of TGFβ1+CCR5+ neutrophil numbers in BM could treat or prevent age-related osteoporosis.
Collapse
Affiliation(s)
- Jinbo Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Institute of Health and Medical Research, Hebei Medical University, Shijiazhuang, Hebei, 050017, China.
| | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Xin Liu
- Department of Orthopedics, Tianjin Hospital, Tianjin, China
| | - Rong Duan
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Xiangjiao Yi
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Akram Ayoub
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Leica Biosystems, Deer Park, IL, 60010, USA
| | - James O Sanders
- Department of Orthopaedics and Rehabilitation Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of North Carolina, Chapel Hill, NC, 27514, USA
| | - Addisu Mesfin
- Department of Orthopaedics and Rehabilitation Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Department of Orthopaedics and Rehabilitation Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
22
|
Jung J, Gokhale S, Xie P. TRAF3: A novel regulator of mitochondrial physiology and metabolic pathways in B lymphocytes. Front Oncol 2023; 13:1081253. [PMID: 36776285 PMCID: PMC9911533 DOI: 10.3389/fonc.2023.1081253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Mitochondria, the organelle critical for cell survival and metabolism, are exploited by cancer cells and provide an important therapeutic target in cancers. Mitochondria dynamically undergo fission and fusion to maintain their diverse functions. Proteins controlling mitochondrial fission and fusion have been recognized as essential regulators of mitochondrial functions, mitochondrial quality control, and cell survival. In a recent proteomic study, we identified the key mitochondrial fission factor, MFF, as a new interacting protein of TRAF3, a known tumor suppressor of multiple myeloma and other B cell malignancies. This interaction recruits the majority of cytoplasmic TRAF3 to mitochondria, allowing TRAF3 to regulate mitochondrial morphology, mitochondrial functions, and mitochondria-dependent apoptosis in resting B lymphocytes. Interestingly, recent transcriptomic, metabolic and lipidomic studies have revealed that TRAF3 also vitally regulates multiple metabolic pathways in B cells, including phospholipid metabolism, glucose metabolism, and ribonucleotide metabolism. Thus, TRAF3 emerges as a novel regulator of mitochondrial physiology and metabolic pathways in B lymphocytes and B cell malignancies. Here we review current knowledge in this area and discuss relevant clinical implications.
Collapse
Affiliation(s)
- Jaeyong Jung
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
23
|
Zeng C, Wang S, Chen F, Wang Z, Li J, Xie Z, Ma M, Wang P, Shen H, Wu Y. Alpinetin alleviates osteoporosis by promoting osteogenic differentiation in BMSCs by triggering autophagy via PKA/mTOR/ULK1 signaling. Phytother Res 2023; 37:252-270. [PMID: 36104214 PMCID: PMC10087978 DOI: 10.1002/ptr.7610] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 01/19/2023]
Abstract
Osteoporosis, a systemic bone disease that is characterized by a reduction in bone mass and destruction of bone microstructure, is becoming a serious problem worldwide. Bone marrow mesenchymal stem cells (BMSCs) can differentiate into bone-forming osteoblasts, and play an important role in maintaining homeostasis of bone metabolism, thus being a potential therapeutic target for osteoporosis. Although the phytochemical alpinetin (APT) has been reported to possess a variety of pharmacological activities, it is still unclear whether APT can influence the osteogenic differentiation of on BMSCs and if it can improve osteoporosis. In this study, we found that APT treatment was able to enhance osteogenic differentiation levels of human BMSCs in vitro and mouse ones in vivo as revealed by multiple osteogenic markers including increased alkaline phosphatase activity and osteocalcin expression. Mechanistically, the protein kinase A (PKA)/mTOR/ULK1 signaling was involved in the action of APT to enhance the osteogenic differentiation of BMSCs. In addition, oral administration of APT significantly mitigated the bone loss in a dexamethasone-induced mouse model of osteoporosis through strengthening PKA signaling and autophagy. Altogether, these data demonstrate that APT promotes osteogenic differentiation in BMSCs by augmenting the PKA/mTOR/ULK1 autophagy signaling, highlighting its potential therapeutic application for treating osteoporotic diseases.
Collapse
Affiliation(s)
- Chenying Zeng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Shan Wang
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Fenglei Chen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Ziming Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Jinteng Li
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Zhongyu Xie
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Mengjun Ma
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Peng Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Huiyong Shen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanfeng Wu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| |
Collapse
|
24
|
Qi G, Jiang Z, Lu W, Li D, Chen W, Yang X, Ding L, Yuan H. Berbamine inhibits RANKL- and M-CSF-mediated osteoclastogenesis and alleviates ovariectomy-induced bone loss. Front Pharmacol 2022; 13:1032866. [PMID: 36408260 PMCID: PMC9666778 DOI: 10.3389/fphar.2022.1032866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/21/2022] [Indexed: 10/06/2023] Open
Abstract
Osteoporosis is a common public health problem characterized by decreased bone mass, increased bone brittleness and damage to the bone microstructure. Excessive bone resorption by osteoclasts is the main target of the currently used drugs or treatment for osteoporosis. Effective antiresorptive drugs without side effects following long-term administration have become a major focus of anti-osteoporotic drugs. In the present study, we investigated the effect of berbamine, a small molecule natural product from Berberis amurensis Rupr, a traditional Chinese medicine, on RANKL-induced osteoclast differentiation in vitro and ovariectomy-induced bone loss in vivo. The results demonstrated that berbamine at a safe and effective dose inhibited osteoclastogenesis and bone resorption function in vitro by suppressing the nuclear factor-κB signaling pathway. In addition, berbamine protected against osteoporosis by inhibiting osteoclastogenesis and bone resorption function without affecting osteogenesis in the ovariectomy mouse model. These findings revealed that berbamine has a protective role against osteoporosis and may represent a novel promising treatment strategy for osteoporosis.
Collapse
Affiliation(s)
- Guobin Qi
- Department of Orthopaedics, Shanghai Sixth People’s Hospital, Shanghai, China
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zengxin Jiang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital, Shanghai, China
| | - Wei Lu
- Department of Orthopedic Surgery, Shanghai TCM-Integrated Hospital Shanghai University of TCM, Shanghai, China
| | - Defang Li
- Department of Orthopedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China
| | - Weibing Chen
- Department of Orthopedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiuying Yang
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Lei Ding
- Department of Orthopedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China
| | - Hengfeng Yuan
- Department of Orthopaedics, Shanghai Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
25
|
Yao Z, Boyce BF. TGFβ priming promotes TNF-induced bone erosion: a promising new target in RA? Nat Rev Rheumatol 2022; 18:617-618. [PMID: 36114427 DOI: 10.1038/s41584-022-00843-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Zhenqiang Yao
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Brendan F Boyce
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
26
|
Tudorancea IM, Ciorpac M, Stanciu GD, Caratașu C, Săcărescu A, Ignat B, Burlui A, Rezuș E, Creangă I, Alexa-Stratulat T, Tudorancea I, Tamba BI. The Therapeutic Potential of the Endocannabinoid System in Age-Related Diseases. Biomedicines 2022; 10:2492. [PMID: 36289755 PMCID: PMC9599275 DOI: 10.3390/biomedicines10102492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/24/2022] [Accepted: 10/03/2022] [Indexed: 11/25/2022] Open
Abstract
The endocannabinoid system (ECS) dynamically regulates many aspects of mammalian physiology. ECS has gained substantial interest since growing evidence suggests that it also plays a major role in several pathophysiological conditions due to its ability to modulate various underlying mechanisms. Furthermore, cannabinoids, as components of the cannabinoid system (CS), have proven beneficial effects such as anti-inflammatory, immunomodulatory, neuromodulatory, antioxidative, and cardioprotective effects. In this comprehensive review, we aimed to describe the complex interaction between CS and most common age-related diseases such as neuro-degenerative, oncological, skeletal, and cardiovascular disorders, together with the potential of various cannabinoids to ameliorate the progression of these disorders. Since chronic inflammation is postulated as the pillar of all the above-mentioned medical conditions, we also discuss in this paper the potential of CS to ameliorate aging-associated immune system dysregulation.
Collapse
Affiliation(s)
- Ivona Maria Tudorancea
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
| | - Mitică Ciorpac
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
| | - Gabriela Dumitrița Stanciu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
| | - Cătălin Caratașu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
| | - Alina Săcărescu
- Department of Medical Specialties II, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității, 700115 Iași, Romania
- Department of Neurology, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa, 700661 Iași, Romania
| | - Bogdan Ignat
- Department of Neurology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alexandra Burlui
- Department of Rheumatology and Rehabilitation, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Elena Rezuș
- Department of Rheumatology and Rehabilitation, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Ioana Creangă
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
- Oncology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Teodora Alexa-Stratulat
- Oncology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Oncology Department, Regional Institute of Oncology, 700483 Iași, Romania
| | - Ionuț Tudorancea
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Cardiology Clinic “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Bogdan Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
- Department of Pharmacology, Clinical Pharmacology and Algesiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
| |
Collapse
|
27
|
Huang X, Lan Y, Shen J, Chen Z, Xie Z. Extracellular Vesicles in Bone Homeostasis: Emerging Mediators of Osteoimmune Interactions and Promising Therapeutic Targets. Int J Biol Sci 2022; 18:4088-4100. [PMID: 35844790 PMCID: PMC9274499 DOI: 10.7150/ijbs.69816] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/27/2022] [Indexed: 11/16/2022] Open
Abstract
An imbalance in bone homeostasis results in bone loss and poor healing in bone diseases and trauma. Osteoimmune interactions, as a key contributor to bone homeostasis, depend on the crosstalk between mesenchymal stem cell-osteoblast (MSC-OB) and monocyte-macrophage (MC-Mφ) lineages. Currently, extracellular vesicles (EVs) are considered to be involved in cell-to-cell communication and represent a novel avenue to enhance our understanding of bone homeostasis and to develop novel diagnostic and therapeutic options. In this comprehensive review, we aim to present recent advances in the study of the effect of MC-Mφ-derived EVs on osteogenesis and the regulatory effects of MSC-OB-derived EVs on the differentiation, recruitment and efferocytosis of Mφ. Furthermore, we discuss the role of EVs as crucial mediators of the communication between these cell lineages involved in the development of common bone diseases, with a focus on osteoporosis, osteoarthritis, bone fracture, and periodontal disease. Together, this review focuses on the apparent discrepancies in current research findings and future directions for translating fundamental insights into clinically relevant EV-based therapies for improving bone health.
Collapse
Affiliation(s)
- Xiaoyuan Huang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Yanhua Lan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Jiahui Shen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Zhuo Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Zhijian Xie
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| |
Collapse
|
28
|
Bolamperti S, Villa I, Rubinacci A. Bone remodeling: an operational process ensuring survival and bone mechanical competence. Bone Res 2022; 10:48. [PMID: 35851054 PMCID: PMC9293977 DOI: 10.1038/s41413-022-00219-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 05/02/2022] [Accepted: 05/15/2022] [Indexed: 12/12/2022] Open
Abstract
Bone remodeling replaces old and damaged bone with new bone through a sequence of cellular events occurring on the same surface without any change in bone shape. It was initially thought that the basic multicellular unit (BMU) responsible for bone remodeling consists of osteoclasts and osteoblasts functioning through a hierarchical sequence of events organized into distinct stages. However, recent discoveries have indicated that all bone cells participate in BMU formation by interacting both simultaneously and at different differentiation stages with their progenitors, other cells, and bone matrix constituents. Therefore, bone remodeling is currently considered a physiological outcome of continuous cellular operational processes optimized to confer a survival advantage. Bone remodeling defines the primary activities that BMUs need to perform to renew successfully bone structural units. Hence, this review summarizes the current understanding of bone remodeling and future research directions with the aim of providing a clinically relevant biological background with which to identify targets for therapeutic strategies in osteoporosis.
Collapse
Affiliation(s)
- Simona Bolamperti
- Osteoporosis and Bone and Mineral Metabolism Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milano, Italy
| | - Isabella Villa
- Osteoporosis and Bone and Mineral Metabolism Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milano, Italy
| | - Alessandro Rubinacci
- Osteoporosis and Bone and Mineral Metabolism Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milano, Italy.
| |
Collapse
|
29
|
Cai P, Yan S, Lu Y, Zhou X, Wang X, Wang M, Yin Z. Carnosol inhibits osteoclastogenesis in vivo and in vitro by blocking the RANKL‑induced NF‑κB signaling pathway. Mol Med Rep 2022; 26:225. [PMID: 35593299 PMCID: PMC9178676 DOI: 10.3892/mmr.2022.12741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Bone homeostasis is maintained by osteoclast-mediated bone resorption and osteoblast‑mediated bone formation. Disruption of bone homeostasis due to excessive osteoclastogenesis or reduced osteogenesis results in various disorders, such as postmenopausal osteoporosis. Receptor activator of NF‑κB ligand (RANKL) stimulation of the NF‑κB signaling pathway is essential in osteoclastogenesis. The aim of the present study was to investigate the novel effects of carnosol, an active compound found in Rosmarinus officinalis, on RANKL‑induced osteoclastogenesis both in vitro and in vivo. TRAP staining showed that carnosol significantly inhibited osteoclasts differentiation of bone marrow monocytes and RAW264.7 cells. Western blot results showed that the protein expression levels of osteoclastogenesis‑associated genes, including cathepsin K, tartrate‑resistant acid phosphatase and MMP‑9, were markedly inhibited by carnosol, which may have suppressed osteoclast function. Furthermore, western blot and immunofluorescent staining results revealed that carnosol markedly suppressed the phosphorylation of p65 induced by RANKL and blocked its nuclear translocation, indicating the suppression of NF‑κB signaling pathway. H&E staining and micro‑CT results showed that in vivo treatment with carnosol significantly attenuated ovariectomy‑induced bone loss in mice. In conclusion, the present study indicated that carnosol may suppress osteoclastogenesis both in vivo and in vitro by inhibiting the activation of the NF‑κB signaling pathway. Carnosol may therefore be a potential novel therapeutic candidate for the clinical treatment of osteoclast‑related disorders.
Collapse
Affiliation(s)
- Pan Cai
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
| | - Shichang Yan
- Department of Orthopedics, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, Jiangsu 210019, P.R. China
| | - Yan Lu
- Department of Laboratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
| | - Xiaoxiao Zhou
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
| | - Xiuhui Wang
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
| | - Minghui Wang
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, P.R. China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, P.R. China
| |
Collapse
|
30
|
Han X, Ren J, Lohner H, Yakoumatos L, Liang R, Wang H. SGK1 negatively regulates inflammatory immune responses and protects against alveolar bone loss through modulation of TRAF3 activity. J Biol Chem 2022; 298:102036. [PMID: 35588785 PMCID: PMC9190018 DOI: 10.1016/j.jbc.2022.102036] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 12/05/2022] Open
Abstract
Serum- and glucocorticoid-regulated kinase 1 (SGK1) is a serine/threonine kinase that plays important roles in the cellular stress response. While SGK1 has been reported to restrain inflammatory immune responses, the molecular mechanisms involved remain elusive, especially in oral bacteria-induced inflammatory milieu. Here, we found that SGK1 curtails Porphyromonas gingivalis-induced inflammatory responses through maintaining levels of tumor necrosis factor receptor-associated factor (TRAF) 3, thereby suppressing NF-κB signaling. Specifically, SGK1 inhibition significantly enhances production of proinflammatory cytokines, including tumor necrosis factor α, interleukin (IL)-6, IL-1β, and IL-8 in P. gingivalis-stimulated innate immune cells. The results were confirmed with siRNA and LysM-Cre-mediated SGK1 KO mice. Moreover, SGK1 deletion robustly increased NF-κB activity and c-Jun expression but failed to alter the activation of mitogen-activated protein kinase signaling pathways. Further mechanistic data revealed that SGK1 deletion elevates TRAF2 phosphorylation, leading to TRAF3 degradation in a proteasome-dependent manner. Importantly, siRNA-mediated traf3 silencing or c-Jun overexpression mimics the effect of SGK1 inhibition on P. gingivalis-induced inflammatory cytokines and NF-κB activation. In addition, using a P. gingivalis infection-induced periodontal bone loss model, we found that SGK1 inhibition modulates TRAF3 and c-Jun expression, aggravates inflammatory responses in gingival tissues, and exacerbates alveolar bone loss. Altogether, we demonstrated for the first time that SGK1 acts as a rheostat to limit P. gingivalis-induced inflammatory immune responses and mapped out a novel SGK1-TRAF2/3-c-Jun-NF-κB signaling axis. These findings provide novel insights into the anti-inflammatory molecular mechanisms of SGK1 and suggest novel interventional targets to inflammatory diseases relevant beyond the oral cavity.
Collapse
Affiliation(s)
- Xiao Han
- Department of Oral and Craniofacial Molecular Biology, VCU Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Junling Ren
- Department of Oral and Craniofacial Molecular Biology, VCU Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Hannah Lohner
- Department of Oral and Craniofacial Molecular Biology, VCU Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Lan Yakoumatos
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Ruqiang Liang
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, California, USA
| | - Huizhi Wang
- Department of Oral and Craniofacial Molecular Biology, VCU Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, USA.
| |
Collapse
|
31
|
Wang T. Searching for the link between inflammaging and sarcopenia. Ageing Res Rev 2022; 77:101611. [PMID: 35307560 DOI: 10.1016/j.arr.2022.101611] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/14/2022] [Accepted: 03/15/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Tiantian Wang
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
32
|
Liu J, Zhang J, Lin X, Boyce BF, Zhang H, Xing L. Age-associated callus senescent cells produce TGF-β1 that inhibits fracture healing in aged mice. J Clin Invest 2022; 132:e148073. [PMID: 35426372 PMCID: PMC9012290 DOI: 10.1172/jci148073] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 02/16/2022] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence plays an important role in human diseases, including osteoporosis and osteoarthritis. Senescent cells (SCs) produce the senescence-associated secretory phenotype to affect the function of neighboring cells and SCs themselves. Delayed fracture healing is common in the elderly and is accompanied by reduced mesenchymal progenitor cells (MPCs). However, the contribution of cellular senescence to fracture healing in the aged has not to our knowledge been studied. Here, we used C57BL/6J 4-month-old young and 20-month-old aged mice and demonstrated a rapid increase in SCs in the fracture callus of aged mice. The senolytic drugs dasatinib plus quercetin enhanced fracture healing in aged mice. Aged callus SCs inhibited the growth and proliferation of callus-derived MPCs (CaMPCs) and expressed high levels of TGF-β1. TGF-β-neutralizing Ab prevented the inhibitory effects of aged callus SCs on CaMPCs and promoted fracture healing in aged mice, which was associated with increased CaMPCs and proliferating cells. Thus, fracture triggered a significant cellular senescence in the callus cells of aged mice, which inhibited MPCs by expressing TGF-β1. Short-term administration of dasatinib plus quercetin depleted callus SCs and accelerated fracture healing in aged mice. Senolytic drugs represent a promising therapy, while TGF-β1 signaling is a molecular mechanism for fractures in the elderly via SCs.
Collapse
Affiliation(s)
- Jiatong Liu
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Jun Zhang
- Plastic Surgery Center, Department of Orthopedics, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Xi Lin
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Brendan F. Boyce
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Hengwei Zhang
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
33
|
Xiao M, Qian H, Lv J, Wang P. Advances in the Study of the Mechanisms of Physiological Root Resorption in Deciduous Teeth. Front Pediatr 2022; 10:850826. [PMID: 35433548 PMCID: PMC9005890 DOI: 10.3389/fped.2022.850826] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/23/2022] [Indexed: 11/15/2022] Open
Abstract
Physiological root resorption of deciduous teeth is a complex physiological process that is essential for the normal replacement of deciduous teeth and permanent teeth in clinical practice, but its importance is often overlooked due to the presence of permanent teeth. This physiological process includes not only the resorption of hard tissues of deciduous teeth, such as dentin and cementum, but also the elimination of soft tissues, such as pulp and periodontal ligament (PDL). However, the mechanisms of physiological root resorption are not yet clear. In this article, the advances of research on the mechanisms related to physiological root resorption will be reviewed in two main aspects: hard tissues and soft tissues of deciduous teeth, specifically in relation to the effects of inflammatory microenvironment and mechanical stress on the resorption of hard tissues, the repair of hard tissues, and the elimination and the histological events of soft tissues.
Collapse
Affiliation(s)
- Manxue Xiao
- Department of Pediatric Dentistry, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Hong Qian
- Department of Pediatric Dentistry, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Jingwen Lv
- Department of Pediatric Dentistry, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Peixuan Wang
- Department of Pediatric Dentistry, Stomatological Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
34
|
Regulation of TNF-Induced Osteoclast Differentiation. Cells 2021; 11:cells11010132. [PMID: 35011694 PMCID: PMC8750957 DOI: 10.3390/cells11010132] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/27/2022] Open
Abstract
Increased osteoclast (OC) differentiation and activity is the critical event that results in bone loss and joint destruction in common pathological bone conditions, such as osteoporosis and rheumatoid arthritis (RA). RANKL and its decoy receptor, osteoprotegerin (OPG), control OC differentiation and activity. However, there is a specific concern of a rebound effect of denosumab discontinuation in treating osteoporosis. TNFα can induce OC differentiation that is independent of the RANKL/RANK system. In this review, we discuss the factors that negatively and positively regulate TNFα induction of OC formation, and the mechanisms involved to inform the design of new anti-resorptive agents for the treatment of bone conditions with enhanced OC formation. Similar to, and being independent of, RANKL, TNFα recruits TNF receptor-associated factors (TRAFs) to sequentially activate transcriptional factors NF-κB p50 and p52, followed by c-Fos, and then NFATc1 to induce OC differentiation. However, induction of OC formation by TNFα alone is very limited, since it also induces many inhibitory proteins, such as TRAF3, p100, IRF8, and RBP-j. TNFα induction of OC differentiation is, however, versatile, and Interleukin-1 or TGFβ1 can enhance TNFα-induced OC formation through a mechanism which is independent of RANKL, TRAF6, and/or NF-κB. However, TNFα polarized macrophages also produce anabolic factors, including insulin such as 6 peptide and Jagged1, to slow down bone loss in the pathological conditions. Thus, the development of novel approaches targeting TNFα signaling should focus on its downstream molecules that do not affect its anabolic effect.
Collapse
|
35
|
Shu-Di-Huang and Gan-Cao Herb Pair Restored the Differentiation Potentials of Mesenchymal Stem Progenitors in Treating Osteoporosis via Downregulation of NF- κB Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:7795527. [PMID: 34950216 PMCID: PMC8692010 DOI: 10.1155/2021/7795527] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/19/2021] [Indexed: 12/20/2022]
Abstract
Background Shu-Di-Huang (Radix Rehmanniae Praeparata, RR) and Gan-Cao (liquorice, L) are frequently used traditional Chinese herb pair in treating osteoporosis (OP). However, the exact mechanism of the RR and L herb pair (RR-L) remains unclear. To explore the efficacy and possible mechanisms of RR-L in treating OP, in silico, in vitro, and in vivo experiments were conducted in the current study. Methods In silico, potential therapeutic target genes and active chemical compounds of RR-L herb pair were predicted and constructed into a network. In vivo, 30 Sprague Dawley rats were divided into 3 groups, including the sham group, the OP model group, and the RR-L-treated OP group. Micro-CT and pathological sections were conducted to validate the therapeutic effects of RR-L in treating OP. MSCs of rats were isolated and cultured in vitro to validate the mesenchymal stem cells (MSCs) related phenotype changes, including Alizarin red staining, Oil red staining, and immunofluorescence. In vitro, cell proliferation analysis, Alizarin red staining, Oil red staining, immunofluorescence of NF-κB, and protein expression of PPARγ, RUNX2, OCN, and p65 were conducted on MSCs to explore the RR-L containing serum in vitro. Also, activator and inhibitor of NF-κB signaling pathway were introduced to determine the possible mechanism of RR-L in the treatment of OP via enhancing MSCs proliferation and differentiation. Results In silico, 168 chemical compounds with a property of oral bioavailability ≥30% and drug-likeness ≥0.18 were recognized as potentially active compounds in RR-L and 249 genes were found to be the targets of which. Among them, 120 genes were found to be therapeutic genes of RR-L in treating OP and KEGG and GO analysis of which demonstrated that RR-L involves in lipid metabolism and multiple inflammation-related signaling pathways. In vivo, ovariectomy- (OVX-) induced OP phenotypes in Sprague Dawley rats include bone mineral density and microarchitecture damaging, abnormal bone metabolism, upregulation of inflammation markers, and damaged differentiation potential of MSCs. Treatment of RR-L reversed the trend and restored the differentiation potential of MSCs. In vitro, RR-L containing serum promoted the osteogenic differentiation and suppressed adipogenic differentiation of MSCs via downregulation of the NF-κB signaling pathway. Also, RR-L containing serum inhibited the tumor necrosis factor-α (TNF-α) induced activation of the NF-κB signaling pathway. On the opposite, the addition of the NF-κB specific inhibitor significantly reduced the effect of RR-L on MSCs. Conclusions In the current study, network pharmacology prediction and experimental validation elucidated that the RR-L herb pair restored damaged MSC differentiation potential via the NF-κB signaling pathway; this could be the possible mechanism of RR-L in treating OP. This finding provides an alternative option in OP therapy.
Collapse
|
36
|
Yang Y, Yang M, Shi D, Chen K, Zhao J, He S, Bai Y, Shen P, Ni H. Single-cell RNA Seq reveals cellular landscape-specific characteristics and potential etiologies for adolescent idiopathic scoliosis. JOR Spine 2021; 4:e1184. [PMID: 35005449 PMCID: PMC8717101 DOI: 10.1002/jsp2.1184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/22/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUNDS Abnormal vertebral growth and development have been found in adolescent idiopathic scoliosis (AIS) patients, and the proliferation and differentiation of bone development-related cells play important roles in its pathogenesis. However, a comprehensive single-cell-level differentiation roadmap in AIS has not been achieved. METHODS The present study compared the single-cell level cellular landscapes of spinal cancellous bone tissues between AIS patients and healthy subjects using high throughput single-cell RNA sequencing (scRNA-seq), which covers multiple cellular lineages including osteoblast, chondrocyte, osteoclast and related immunocytes. We constructed the differentiation trajectories of bone development-related cell lineages through pseudotime analysis, and the intercellular-communication networks between bone development-related cells and immunocytes were further developed. RESULTS A total of 11 distinct cell clusters were identified according to the genome-wide transcriptome profiles. t-Distributed stochastic neighbor embedding (t-SNE) analysis showed that mesenchymal stem cells (MSC) were classified into three subtypes: MSC-LOXL2, MSC-IGFBP5, and MSC-GJA1. Gene ontology (GO) analysis showed that MSC-GJA1 might possess greater osteoblast differentiation potential than the others. MSC-IGFBP5 was the specific MSC subtype observed only in AIS. There were two distinct gene expression clusters: OB-DPT and OB-OLFML2B, and the counts of osteoblasts derived from AIS was significantly less than that of non-AIS subjects. In AIS patients, MSC-IGFBP5 failed to differentiate into osteoblasts and exhibited negative regulation of cell proliferation and enhanced cell death. CPC-PCNA was found to be the specific chondrocyte progenitor cell (CPC) subtype observed only in AIS patients. The cell counts of OC-BIRC3 in AIS were less than those in controls. Pseudotime analysis suggested two possible distinct osteoclast differentiation patterns in AIS and control subjects. Monocytes in AIS mainly differentiated into OC-CRISP3. CONCLUSIONS Our single-cell analysis first revealed differences existed in the cellular states between AIS patients and healthy subjects and found the differentiation disruption of specific MSC and CPC clusters in AIS. Cell communication analysis provided the possible pathogenesis of osteoblast and chondrocyte differentiation dysfunction in AIS.
Collapse
Affiliation(s)
- Yilin Yang
- Department of Orthopaedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Mingyuan Yang
- Department of OrthopaedicsChanghai Hospital, Navy Medical UniversityShanghaiChina
| | - Dongliang Shi
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Tongji University School of MedicineShanghaiChina
| | - Kai Chen
- Department of OrthopaedicsChanghai Hospital, Navy Medical UniversityShanghaiChina
| | - Jian Zhao
- Department of OrthopaedicsChanghai Hospital, Navy Medical UniversityShanghaiChina
| | - Shisheng He
- Department of Orthopaedics, Shanghai 10th People's HospitalTongji UniversityShanghaiChina
| | - Yushu Bai
- Department of OrthopaedicsChanghai Hospital, Navy Medical UniversityShanghaiChina
| | - Pinquan Shen
- Department of Pediatric Orthopaedics, Xinhua HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Haijian Ni
- Department of Orthopaedics, Shanghai 10th People's HospitalTongji UniversityShanghaiChina
| |
Collapse
|
37
|
Yi X, Liu X, Kenney HM, Duan R, Lin X, Schwarz E, Yao Z. TNF-Polarized Macrophages Produce Insulin-like 6 Peptide to Stimulate Bone Formation in Rheumatoid Arthritis in Mice. J Bone Miner Res 2021; 36:2426-2439. [PMID: 34585777 PMCID: PMC8688308 DOI: 10.1002/jbmr.4447] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/01/2021] [Accepted: 09/22/2021] [Indexed: 01/05/2023]
Abstract
The risk of osteoporosis is increased in rheumatoid arthritis (RA). Anti-tumor necrosis factor (TNF) therapy has markedly improved the outcomes of RA patients but does not improve osteoporosis in some reports. This could be a combined result of disease severity and other therapeutic agents, such as glucocorticoids that accelerate osteoporosis progression. We evaluated the effects of anti-TNF therapy on osteoporosis in an animal model of RA and explored the possible mechanisms involved. Six-week-old TNF transgenic (TNF-Tg) mice with early stage erosive arthritis were treated with TNF antibody (Ab) or control immunoglobulin (IgG) weekly for 4 weeks. We found that TNF Ab completely blocked the development of erosive arthritis in TNF-Tg mice, but only slightly increased vertebral bone mass, associated with reduction in parameters of both bone resorption and formation. Similarly, TNF Ab slightly increased trabecular bone mass in tibias of 8-month-old TNF-Tg mice with advanced erosive arthritis. Interestingly, TNFα increased osteoblast differentiation from mouse bone marrow stromal cells (BMSCs) containing large number of macrophages but not from pure mesenchymal progenitor cells (MPCs). TNFα-polarized macrophages (TPMs) did not express iNos and Arginase 1, typical markers of inflammatory and resident macrophages. Interestingly, TPMs stimulated osteoblast differentiation, unlike resident and inflammatory macrophages polarized by IL-4 and interferon-λ, respectively. RNA-seq analysis indicated that TPMs produced several anabolic factors, including Jagged1 and insulin like 6 (INSL6). Importantly, inhibition of either Jagged1 or INSL6 blocked TNFα-induced osteoblast differentiation. Furthermore, INSL6 Ab significantly decreased the expansion of TNF-induced MPCs in BMSCs, and anti-TNF Ab reduced INSL6 expression by macrophages in vitro and in TNF-Tg mice in vivo. We conclude that TPMs produce INSL6 to stimulate bone formation and anti-TNF Ab blocks not only enhanced bone resorption but also the anabolic effect of TPMs on bone, limiting its effect to increase bone mass in this model of RA. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Xiangjiao Yi
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Xin Liu
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - H Mark Kenney
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Rong Duan
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Xi Lin
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward Schwarz
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopedic Surgery, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
38
|
Zhou Y, Deng Y, Liu Z, Yin M, Hou M, Zhao Z, Zhou X, Yin L. Cytokine-scavenging nanodecoys reconstruct osteoclast/osteoblast balance toward the treatment of postmenopausal osteoporosis. SCIENCE ADVANCES 2021; 7:eabl6432. [PMID: 34818042 PMCID: PMC8612675 DOI: 10.1126/sciadv.abl6432] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Imbalance between osteoblasts and osteoclasts accounts for the incidence and deterioration of postmenopausal osteoporosis. Abnormally elevated RANKL and TNF-α levels after menopause promote osteoclast formation and inhibit osteoblast differentiation, respectively. Here, nanodecoys capable of scavenging RANKL and TNF-α were developed from preosteoclast (RAW 264.7 cell) membrane–coated poly(lactic-co-glycolic acid) (PLGA) nanoparticles, which inhibited osteoporosis and maintained bone integrity. The nanodecoys effectively escaped from macrophage capture and enabled prolonged blood circulation after systemic administration. The abundant RANK and TNF-α receptor (TNF-αR) on the cell membranes effectively neutralized RANKL and TNF-α to prevent osteoclastogenesis and promote osteoblastogenesis, respectively, thus reversing the progression of osteoporosis in the ovariectomized (OVX) mouse model. These biomimetic nanodecoys provide an effective strategy for reconstructing the osteoclast/osteoblast balance and hold great potentials for the clinical management of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Yang Zhou
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yekun Deng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Zhongmin Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Mengyuan Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Mengying Hou
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Ziyin Zhao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Xiaozhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- Corresponding author.
| |
Collapse
|
39
|
Bellavia D, Caradonna F, Dimarco E, Costa V, Carina V, De Luca A, Raimondi L, Gentile C, Alessandro R, Fini M, Giavaresi G. Terpenoid treatment in osteoporosis: this is where we have come in research. Trends Endocrinol Metab 2021; 32:846-861. [PMID: 34481733 DOI: 10.1016/j.tem.2021.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 01/30/2023]
Abstract
Lower bone resistance to load is due to the imbalance of bone homeostasis, where excessive bone resorption, compared with bone formation, determines a progressive osteopenia, leading to a high risk of fractures and consequent pain and functional limitations. Terpenoids, with their activities against bone resorption, have recently received increased attention from researchers. They are potentially more suitable for long-term use compared with traditional therapeutics. In this review of the literature of the past 5 years, we provide comprehensive information on terpenoids, with their anti-osteoporotic effects, highlighting molecular mechanisms that are often in epigenetic key and a possible pharmacological use in osteoporosis prevention and treatment.
Collapse
Affiliation(s)
- Daniele Bellavia
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Bologna, Italy.
| | - Fabio Caradonna
- University of Palermo, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Section of Cellular Biology, Palermo, Italy
| | - Eufrosina Dimarco
- University of Palermo, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Section of Cellular Biology, Palermo, Italy
| | - Viviana Costa
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Bologna, Italy
| | - Valeria Carina
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Bologna, Italy
| | - Angela De Luca
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Bologna, Italy
| | - Lavinia Raimondi
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Bologna, Italy
| | - Carla Gentile
- University of Palermo, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Section of Cellular Biology, Palermo, Italy
| | - Riccardo Alessandro
- University of Palermo, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Biology and Genetics, Palermo, Italy; Istituto per la Ricerca e l'Innovazione Biomedica (IRIB), Consiglio Nazionale delle Ricerche (CNR), Palermo, Italy
| | - Milena Fini
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Bologna, Italy
| | - Gianluca Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, SC Scienze e Tecnologie Chirurgiche - SS Piattaforma Scienze Omiche per Ortopedia Personalizzata, Bologna, Italy
| |
Collapse
|
40
|
Li Z, Xue H, Tan G, Xu Z. Effects of miRNAs, lncRNAs and circRNAs on osteoporosis as regulatory factors of bone homeostasis (Review). Mol Med Rep 2021; 24:788. [PMID: 34505632 PMCID: PMC8441966 DOI: 10.3892/mmr.2021.12428] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/25/2021] [Indexed: 01/03/2023] Open
Abstract
Osteoporosis is a common metabolic bone disorder typically characterized by decreased bone mass and an increased risk of fracture. At present, the detailed molecular mechanism underlying the development of osteoporosis remains to be elucidated. Accumulating evidence shows that non-coding (nc)RNAs, such as microRNAs (miRNAs), long ncRNAs (lncRNAs) and circular RNAs (circRNAs), play significant roles in osteoporosis through the post-transcriptional regulation of gene expression as regulatory factors. Previous studies have demonstrated that ncRNAs participate in maintaining bone homeostasis by regulating physiological and developmental processes in osteoblasts, osteoclasts and bone marrow stromal cells. In the present review, the latest research investigating the involvement of miRNAs, lncRNAs and circRNAs in regulating the differentiation, proliferation, apoptosis and autophagy of cells that maintain the bone microenvironment in osteoporosis is summarized. Deeper insight into the aspects of osteoporosis pathogenesis involving the deregulation of ncRNAs could facilitate the development of therapeutic approaches for osteoporosis.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Haipeng Xue
- Department of Orthopaedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Guoqing Tan
- Department of Orthopaedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Zhanwang Xu
- Department of Orthopaedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| |
Collapse
|
41
|
So T. The immunological significance of tumor necrosis factor receptor-associated factors (TRAFs). Int Immunol 2021; 34:7-20. [PMID: 34453532 DOI: 10.1093/intimm/dxab058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 07/27/2021] [Indexed: 01/03/2023] Open
Abstract
The tumor necrosis factor receptor (TNFR)-associated factor (TRAF) family of molecules are intracellular signaling adaptors and control diverse signaling pathways mediated not only by the TNFR superfamily and the Toll-like receptor/interleukin-1 receptor superfamily but also by unconventional cytokine receptors such as IL-6 and IL-17 receptors. There are seven family members, TRAF1 to TRAF7, in mammals. Exaggerated immune responses induced through TRAF signaling downstream of these receptors often lead to inflammatory and autoimmune diseases including rheumatoid arthritis, inflammatory bowel disease, psoriasis and autoinflammatory syndromes, and thus those signals are major targets for therapeutic intervention. For this reason, it has been very important to understand signaling mechanisms regulated by TRAFs that greatly impact on life/death decisions and the activation, differentiation and survival of cells of the innate and adaptive immune systems. Accumulating evidence suggests that dysregulated cellular expression and/or signaling of TRAFs causes overproduction of proinflammatory cytokines, which facilitates aberrant activation of immune cells. In this review, I will explain the structural and functional aspects that are responsible for the cellular activity and disease outcomes of TRAFs, and summarize the findings of recent studies on TRAFs in terms of how individual TRAF family molecules regulates biological and disease processes in the body in both positive and negative ways. This review also discusses how TRAF mutations contribute to human disease.
Collapse
Affiliation(s)
- Takanori So
- Laboratory of Molecular Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| |
Collapse
|
42
|
Yang CH, Jeyaprakash N, Tseng YJ. Numerical Simulation and Non-Destructive Characterization of Material Property and Defect Analysis of Cortical Bone Using Laser Ultrasound Techniques. ACS Biomater Sci Eng 2021; 7:3917-3932. [PMID: 34325509 DOI: 10.1021/acsbiomaterials.1c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The application of bone quality assessment has received extensive attention, and a large number of researchers continue to invest in related research activities. To get closer to the real situation, this study intends to investigate the long bones of cattle. A quantitative laser ultrasound visualization (QLUV) system was used to measure the images transmitted by the guided waves on the long bones, and the internal defects of the long bones were detected using wave propagation behavior. Then, linear scanning was performed through a laser ultrasound technique (LUT) to measure the dispersion curve of the cortical bone, and the results were compared with finite element simulations. Further, LUT was used to measure the material properties of the cortical bone in all directions. Finally, the long bones were scanned by computerized tomography to analyze the pore direction and distribution. Further, the relationship between pore direction and material properties was studied. The results showed that the obtained wave propagation image was consistent with the time-domain waveform signal and the finite element analysis results. The experimental and simulation results of wave velocity showed an error of 0.26 to 1.56% compared with the literature. The plate-shaped cortical bone showed that the phase velocity of the guided wave is higher than the circumferential direction. The defect location was identified through wave propagation behavior using the QLUV system. The elastic constant of the cortical bone was measured, and it showed the same trend as the results obtained from the tensile test in the literature. Also, the pore distribution indicated that the cortical bone porosity has the same trend as elastic constants. The elastic constants along the longitudinal direction were greater than the transversal direction. This laser ultrasound technique has been developed with an aim of having a better resolution and also as a potential application in osteoporosis conditions.
Collapse
Affiliation(s)
- Che-Hua Yang
- Additive Manufacturing Center for Mass Customization Production, National Taipei University of Technology, Taipei 10608, Taiwan, ROC.,Graduate Institute of Manufacturing Technology, National Taipei University of Technology, Taipei 10608, Taiwan, ROC
| | - N Jeyaprakash
- Additive Manufacturing Center for Mass Customization Production, National Taipei University of Technology, Taipei 10608, Taiwan, ROC.,Graduate Institute of Manufacturing Technology, National Taipei University of Technology, Taipei 10608, Taiwan, ROC
| | - Yu-Jing Tseng
- Graduate Institute of Manufacturing Technology, National Taipei University of Technology, Taipei 10608, Taiwan, ROC
| |
Collapse
|
43
|
Wang Y, Shen S, Hu T, Williams GR, Bian Y, Feng B, Liang R, Weng X. Layered Double Hydroxide Modified Bone Cement Promoting Osseointegration via Multiple Osteogenic Signal Pathways. ACS NANO 2021; 15:9732-9745. [PMID: 34086438 DOI: 10.1021/acsnano.1c00461] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Poly(methyl methacrylate) (PMMA) bone cement has been widely used in orthopedic surgeries including total hip/knee replacement, vertebral compression fracture treatment, and bone defect filling. However, aseptic loosening of the interface between PMMA bone cement and bone often leads to failure. Hence, the development of modified PMMA that facilitates the growth of bone into the modified PMMA bone cement is key to reducing the incidence of aseptic loosening. In this study, MgAl-layered double hydroxide (LDH) microsheets modified PMMA (PMMA&LDH) bone cement with superior osseointegration performance has been synthesized. The maximum polymerization reaction temperature of PMMA&LDH decreased by 7.0 and 11.8 °C, respectively, compared with that of PMMA and PMMA&COL-I (mineralized collagen I modified PMMA). The mechanical performance of PMMA&LDH decreased slightly in comparison with PMMA, which is beneficial to alleviate stress-shielding osteolysis, and indirectly promote osseointegration. The superior osteogenic ability of PMMA&LDH has been demonstrated in vivo, which boosts bone growth by 2.17- and 18.34-fold increments compared to the PMMA&COL-I and PMMA groups at 2 months, postoperatively. Moreover, transcriptome sequencing revealed four key osteogenic pathways: p38 MAPK, ERK/MAPK, FGF, and TGF-β, which were further confirmed by IPA, qPCR, and Western blot assays. Hence, LDH-modified PMMA bone cement is a promising biomaterial to enhance bone growth with potential applications in relevant orthopedic surgeries.
Collapse
Affiliation(s)
- Yingjie Wang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Songpo Shen
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
- Department of Orthopedic Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Tingting Hu
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, U.K
| | - Yanyan Bian
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Bin Feng
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xisheng Weng
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| |
Collapse
|
44
|
Association of TGF-β1 and IL-10 Gene Polymorphisms with Osteoporosis in a Study of Taiwanese Osteoporotic Patients. Genes (Basel) 2021; 12:genes12060930. [PMID: 34207210 PMCID: PMC8233820 DOI: 10.3390/genes12060930] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 01/07/2023] Open
Abstract
Osteoporosis is a rising health threat in the increasingly aging world population. It is a common skeletal disease strongly linked to genetic predisposition. We aim to identify the effects of the anti-inflammatory TGF-β1- and IL-10-specific single-nucleotide polymorphism (SNP) combination on the risk for osteoporosis. We investigated and analyzed the relationships between three TGF-β1 SNPs (-509C/T, +869 T/C and +29T/C), one IL-10 SNP (+1927A/C) and the level of bone mineral density (BMD), as well as the risk of osteoporosis in Taiwanese osteoporotic patients. A total of 217 subjects were recruited, including 88 osteoporotic patients and 129 healthy controls, for SNPs, BMD and clinical characteristics statistical analyses. Females with TGF-β1 SNP (-509 C/C) and IL-10 SNP (+1927 C/C) genotypes showed a great benefit for femoral neck T-scores. However, the combination of TGF-β1 SNP (-509 T/T) and IL-10 SNP (+1927 A/A) genotypes in all subjects showed a significant decrease in total hip BMD T-scores. The TGF-β1 SNP (-509 C/T) genotype in all subjects and TGF-β1 SNP (-509 T/T) and IL-10 SNP (+1927 A/C) genotypes in males showed positive effects on body height. The combination of the many SNPs in the anti-inflammatory TGF-β1 and IL-10 genes may be cooperatively involved in the development of osteoporosis. Our data suggested that the specific SNP combination of TGF-β1 (-509) and IL-10 (+1927) may act as a predictive factor for postmenopausal osteoporosis in Taiwanese women.
Collapse
|
45
|
Xia H, Liu J, Yang W, Liu M, Luo Y, Yang Z, Xie J, Zeng H, Xu R, Ling H, Zeng Q, Xu H, Fang L, Wang H, Tong P, Jin H, Yang F. Integrated Strategy of Network Pharmacological Prediction and Experimental Validation Elucidate Possible Mechanism of Bu-Yang Herbs in Treating Postmenopausal Osteoporosis via ESR1. Front Pharmacol 2021; 12:654714. [PMID: 34045964 PMCID: PMC8144472 DOI: 10.3389/fphar.2021.654714] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/12/2021] [Indexed: 12/18/2022] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a type of bone metabolism disease-related to estrogen deficiency with an increasing incidence. Traditional Chinese (TCM) has always been used and showed effectiveness in treating PMOP. In the current study, Bu-Yang herbs were considered to be the most frequently used and efficient TCM herbs in PMOP treatment. However, chemical and pharmacological profiles were not elucidated. Network pharmacology was conducted on representative Bu-Yang herbs (Yin-Yang-Huo. Du-Zhong, Bu-Gu-Zhi, Tu-Si-Zi) to investigate the mechanism of Bu-Yang herbs on PMOP. Chemical compounds, potential targets, and disease related genes were available from the corresponding database. Results showed that Bu-Yang herbs could interact with ESR1 and estrogen signaling pathways. For further validation, the Bu-Yang decoction (BYD), formula consisted of the above-mentioned 4 Bu-Yang herbs was presented for experimental validation. In vivo, BYD significantly reversed ovariectomy (OVX)-induced osteoporosis progress in a dose-dependent manner by up-regulation of bone mineral density and amelioration of bone microarchitecture. In vitro, BYD dramatically improved the proliferation and mineral nodules formation of osteoblasts. Both in vitro and in vivo results illustrated that the phenotype change induced by BYD is correlated with up-regulated of ESR1 and activation of the β-catenin pathway. Meanwhile, inhibition of ESR1 by ICI182, 780 blocked the osteogenic phenotype and β-catenin pathway activation induced by BYD. In conclusion, the current study suggested that Bu-Yang herbs are the most useful TCM herbs in treating PMOP. Furthermore, the integrated strategy of network pharmacology prediction with experimental validation suggested that BYD exerted its anti-PMOP via ESR1 and the downstream mechanism might be activation of the β-catenin signaling pathway.
Collapse
Affiliation(s)
- Hanting Xia
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopedics, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Jiangyuan Liu
- Graduated School, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Wenlong Yang
- Department of Orthopedics, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Min Liu
- Department of Orthopedics, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yunfeng Luo
- Graduated School, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Zhijun Yang
- Graduated School, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Jingbo Xie
- Department of Orthopedics, People's Hospital of Fengcheng City, Fengcheng, China
| | - Huiliang Zeng
- Department of Orthopedics, Foshan Hospital of Traditional Chinese Medicine, Foshan, China
| | - Rui Xu
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Houfu Ling
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Qinghe Zeng
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Huihui Xu
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Liang Fang
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongyu Wang
- Graduated School, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Peijian Tong
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongting Jin
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Fengyun Yang
- Graduated School, Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
46
|
Schwab N, Ju Y, Hazrati LN. Early onset senescence and cognitive impairment in a murine model of repeated mTBI. Acta Neuropathol Commun 2021; 9:82. [PMID: 33964983 PMCID: PMC8106230 DOI: 10.1186/s40478-021-01190-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/03/2021] [Indexed: 12/19/2022] Open
Abstract
Mild traumatic brain injury (mTBI) results in broad neurological symptoms and an increased risk of being diagnosed with a neurodegenerative disease later in life. While the immediate oxidative stress response and post-mortem pathology of the injured brain has been well studied, it remains unclear how early pathogenic changes may drive persistent symptoms and confer susceptibility to neurodegeneration. In this study we have used a mouse model of repeated mTBI (rmTBI) to identify early gene expression changes at 24 h or 7 days post-injury (7 dpi). At 24 h post-injury, gene expression of rmTBI mice shows activation of the DNA damage response (DDR) towards double strand DNA breaks, altered calcium and cell–cell signalling, and inhibition of cell death pathways. By 7 dpi, rmTBI mice had a gene expression signature consistent with induction of cellular senescence, activation of neurodegenerative processes, and inhibition of the DDR. At both timepoints gliosis, microgliosis, and axonal damage were evident in the absence of any gross lesion, and by 7 dpi rmTBI also mice had elevated levels of IL1β, p21, 53BP1, DNA2, and p53, supportive of DNA damage-induced cellular senescence. These gene expression changes reflect establishment of processes usually linked to brain aging and suggests that cellular senescence occurs early and most likely prior to the accumulation of toxic proteins. These molecular changes were accompanied by spatial learning and memory deficits in the Morris water maze. To conclude, we have identified DNA damage-induced cellular senescence as a repercussion of repeated mild traumatic brain injury which correlates with cognitive impairment. Pathways involved in senescence may represent viable treatment targets of post-concussive syndrome. Senescence has been proposed to promote neurodegeneration and appears as an effective target to prevent long-term complications of mTBI, such as chronic traumatic encephalopathy and other related neurodegenerative pathologies.
Collapse
|
47
|
Liu H, Yi X, Tu S, Cheng C, Luo J. Kaempferol promotes BMSC osteogenic differentiation and improves osteoporosis by downregulating miR-10a-3p and upregulating CXCL12. Mol Cell Endocrinol 2021; 520:111074. [PMID: 33157164 DOI: 10.1016/j.mce.2020.111074] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Kaempferol has improved the functions of various human diseases. Here, we aimed to probe into the potential molecular mechanism of Kaempferol to ameliorate osteoporosis. METHODS Micro-computed tomography scanning was applied to assess the bone density of osteoporosis rats induced by ovariectomized. Quantitative real-time PCR was applied to detect the expressions of RUNX2, Osterix, CXCL12, and miR-10a-3p. Western blot, Alizarin red staining, Alkaline Phosphatase Diethanolamine Activity Kit were applied to confirm the in vitro functions of Kaempferol. RNA Immunoprecipitation and dual-luciferase reporter gene experiments were applied to study the potential mechanism. RESULTS The treatment of Kaempferol raised bone density in osteoporosis rats induced by ovariectomized, and boosted the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), and raised the expressions of RUNX2, Osterix, and CXCL12, and lessened miR-10a-3p. From the potential mechanism analysis, we corroborated that miR-10a-3p and CXCL12 bound to each other, and Kaempferol boosted BMSC osteogenic differentiation and ameliorated osteoporosis by lessening miR-10a-3p and raising CXCL12. CONCLUSION Our data expounded that Kaempferol boosted BMSC osteogenic differentiation and ameliorated osteoporosis by lessening miR-10a-3p and raising CXCL12.
Collapse
Affiliation(s)
- Hao Liu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xin Yi
- Medical College of Yichun Vocational and Technical College, Yichun, China
| | - ShuTing Tu
- College of Medicine, Nanchang University, Nanchang, China
| | - Chong Cheng
- College of Medicine, Nanchang University, Nanchang, China
| | - Jun Luo
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
48
|
Huang C, Zheng Y, Bai J, Shi C, Shi X, Shan H, Zhou X. Hepatocyte growth factor overexpression promotes osteoclastogenesis and exacerbates bone loss in CIA mice. J Orthop Translat 2020; 27:9-16. [PMID: 33344167 PMCID: PMC7732867 DOI: 10.1016/j.jot.2020.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/11/2020] [Accepted: 10/28/2020] [Indexed: 01/20/2023] Open
Abstract
Background Hepatocyte growth factor (HGF) is a multifunctional growth factor that promotes various biological processes. However, the effect of HGF on bone metabolism in rheumatoid arthritis (RA) remains unknown. Here, we investigated the role of HGF in regulating osteoclastogenesis and bone resorption in RA. Methods The expression of HGF in RA patients and collagen-induced arthritis (CIA) mice was examined. The role of HGF on osteoclastogenesis was analysed by osteoclastogenesis and bone resorption assays. The effect of HGF inhibition was evaluated in a CIA mice model. The mechanism of HGF in regulating osteoclastogenesis and bone resorption was explored by a series of in vitro studies. Results HGF was overexpressed in CIA and RA. HGF stimulated osteoclastogenesis in vitro. SU11274, a selective small molecule blocker of c-Met, impeded the effect of HGF on osteoclastogenesis and bone resorption. HGF regulated osteoclastogenesis by JNK and AKT-GSK-3β-NFATc1 signallings. SU11274 protected CIA mice from pathological bone loss. Conclusions These data strongly suggest that the highly expressed HGF in the joint tissues contributes to bone loss in RA. Inhibition of HGF/c-Met could effectively alleviate pathological bone loss and inflammatory symptoms in CIA mice. HGF/c-Met may be used as a new target for the treatment of bone loss in RA.
Collapse
Affiliation(s)
- Chaoming Huang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China.,Department of Orthopedics, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, Jiangsu, 223800, China
| | - Yufan Zheng
- Institute of Neuroscience, Soochow University, Suzhou, 215000, China
| | - Jinyu Bai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China
| | - Ce Shi
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China.,Department of Orthopedics, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu, 223800, China
| | - Xin Shi
- Institute of Neuroscience, Soochow University, Suzhou, 215000, China
| | - Huajian Shan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China
| | - Xiaozhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China
| |
Collapse
|
49
|
Zhou W, Lin D, Zhong Z, Ye Q. Roles of TRAFs in Ischemia-Reperfusion Injury. Front Cell Dev Biol 2020; 8:586487. [PMID: 33224951 PMCID: PMC7674171 DOI: 10.3389/fcell.2020.586487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor receptor-associated factor (TRAF) proteins are a family of signaling molecules that function downstream of multiple receptor signaling pathways, and they play a pivotal role in the regulation of intracellular biological progresses. These TRAF-dependent signaling pathways and physiological functions have been involved in the occurrence and progression of ischemia-reperfusion injury (IRI), which is a common pathophysiological process that occurs in a wide variety of clinical events, including ischemic shock, organ transplantation, and thrombolytic therapy, resulting in a poor prognosis and high mortality. IRI occurs in multiple organs, including liver, kidney, heart, lung, brain, intestine, and retina. In recent years, mounting compelling evidence has confirmed that the genetic alterations of TRAFs can cause subversive phenotype changes during IRI of those organs. In this review, based on current knowledge, we summarized and analyzed the regulatory effect of TRAFs on the IRI of various organs, providing clear direction and a firm theoretical basis for the development of treatment strategies to manipulate TRAF proteins or TRAF-dependent signaling pathways in IRI-related diseases.
Collapse
Affiliation(s)
- Wei Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Danni Lin
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China.,The First Affiliated Hospital, Zhejiang University School of Medicine, Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Innovation Center for the Study of Pancreatic Diseases, Hangzhou, China
| | - Zibiao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China.,The Third Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, Changsha, China
| |
Collapse
|
50
|
Downregulation of circular RNA HECTD1 induces neuroprotection against ischemic stroke through the microRNA-133b/TRAF3 pathway. Life Sci 2020; 264:118626. [PMID: 33148417 DOI: 10.1016/j.lfs.2020.118626] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 01/16/2023]
Abstract
AIMS Circular RNAs (circRNAs) have been shown to play crucial roles in various biological processes and human diseases. However, their exact functions in ischemic stroke remain largely unknown. In this study, we explored the functional role of circRNA HECTD1 (circ-HECTD1) and its underlying mechanism in cerebral ischemia/reperfusion injury. METHODS Mouse middle cerebral artery occlusion (MCAO) model and oxygen-glucose deprivation (OGD) model in HT22 cells were used to mimic the cerebral ischemia/reperfusion injury. Brain infarct volume, flow cytometry, caspase 3 activity, NF-κB activity, and TUNEL staining were performed to evaluate the function of circ-HECTD1. Luciferase report assay was used to explore the regulatory mechanism. FINDINGS The results showed that the expression of circ-HECTD1 and tumor necrosis factor receptor-associated factor 3 (TRAF3) was remarkably up-regulated, while miR-133b was down-regulated in oxygen-glucose deprivation (OGD)-induced HT22 cells and mouse middle cerebral artery occlusion (MCAO) model. circ-HECTD1 knockdown relieved OGD-caused neuronal cell death in vitro. Simultaneously, circ-HECTD1 knockdown improved cerebral infarction volume and neuronal apoptosis in MCAO mice. circ-HECTD1 was able to negatively regulate the expression of miR-133b, and TRAF3 is one of the targets of miR-133b. Upregulation of miR-133b inhibited the expression of TRAF3 in OGD-stimulated cells, whereas circ-HECTD1 upregulation reversed this effect. Furthermore, upregulation of miR-133 was able to inhibit OGD-caused cell apoptosis and NF-κB activation, whereas upregulation of circ-HECTD1 attenuated these effects of miR-133b mimics. SIGNIFICANCE Taken together, circ-HECTD1 knockdown inhibited the expression of TRAF3 by targeting miR-133b, thereby attenuating neuronal injury caused by cerebral ischemia.
Collapse
|