1
|
Yao Y, Cai X, Chen Y, Zhang M, Zheng C. Estrogen deficiency-mediated osteoimmunity in postmenopausal osteoporosis. Med Res Rev 2024. [PMID: 39234932 DOI: 10.1002/med.22081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/03/2023] [Accepted: 08/25/2024] [Indexed: 09/06/2024]
Abstract
Postmenopausal osteoporosis (PMO) is a common disease associated with aging, and estrogen deficiency is considered to be the main cause of PMO. Recently, however, osteoimmunology has been revealed to be closely related to PMO. On the one hand, estrogen deficiency directly affects the activity of bone cells (osteoblasts, osteoclasts, osteocytes). On the other hand, estrogen deficiency-mediated osteoimmunity also plays a crucial role in bone loss in PMO. In this review, we systematically describe the progress of the mechanisms of bone loss in PMO, estrogen deficiency-mediated osteoimmunity, the differences between PMO patients and postmenopausal populations without osteoporosis, and estrogen deficiency-mediated immune cells (T cells, B cells, macrophages, neutrophils, dendritic cells, and mast cells) activity. The comprehensive summary of this paper provides a clear knowledge context for future research on the mechanism of PMO bone loss.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Xiaoyu Cai
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| |
Collapse
|
2
|
Choi YJ. Cancer treatment-induced bone loss. Korean J Intern Med 2024; 39:731-745. [PMID: 38439172 PMCID: PMC11384245 DOI: 10.3904/kjim.2023.386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/17/2023] [Accepted: 11/27/2023] [Indexed: 03/06/2024] Open
Abstract
Cancer treatment-induced bone loss (CTBL) is associated with anti-tumor treatments, including endocrine therapies, chemotherapeutic treatments, radiotherapy, glucocorticoids, and tyrosine kinase inhibitors. Osteoporosis, characterized by the loss of bone mass, can increase the risk of fractures, leading to mortality and long-term disability, even after cancer remission. Cancer and osteoporosis have marked clinical and pathogenetic similarities. Both have a multifactorial etiology, affect the geriatric population, and markedly influence quality of life. Lifestyle management, including calcium and vitamin D supplementation, is recommended but the supporting evidence is limited. Oral and injectable bisphosphonates are effective for osteoporosis and malignant bone disease. Bisphosphonates increase bone mineral density (BMD) in patients with CTBL. Denosumab is also used in the management of CTBL; in clinical trials, it improved BMD and reduced the risk of fracture. Currently, there are no bone anabolic therapies for patients with cancer. Appropriate therapies are necessary to maintain optimal bone health, particularly in patients at heightened risk.
Collapse
Affiliation(s)
- Yong Jun Choi
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
3
|
Miron RJ, Bohner M, Zhang Y, Bosshardt DD. Osteoinduction and osteoimmunology: Emerging concepts. Periodontol 2000 2024; 94:9-26. [PMID: 37658591 DOI: 10.1111/prd.12519] [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: 05/14/2023] [Revised: 06/23/2023] [Accepted: 07/20/2023] [Indexed: 09/03/2023]
Abstract
The recognition and importance of immune cells during bone regeneration, including around bone biomaterials, has led to the development of an entire field termed "osteoimmunology," which focuses on the connection and interplay between the skeletal system and immune cells. Most studies have focused on the "osteogenic" capacity of various types of bone biomaterials, and much less focus has been placed on immune cells despite being the first cell type in contact with implantable devices. Thus, the amount of literature generated to date on this topic makes it challenging to extract needed information. This review article serves as a guide highlighting advancements made in the field of osteoimmunology emphasizing the role of the osteoimmunomodulatory properties of biomaterials and their impact on osteoinduction. First, the various immune cell types involved in bone biomaterial integration are discussed, including the prominent role of osteal macrophages (OsteoMacs) during bone regeneration. Thereafter, key biomaterial properties, including topography, wettability, surface charge, and adsorption of cytokines, growth factors, ions, and other bioactive molecules, are discussed in terms of their impact on immune responses. These findings highlight and recognize the importance of the immune system and osteoimmunology, leading to a shift in the traditional models used to understand and evaluate biomaterials for bone regeneration.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | | | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| | | |
Collapse
|
4
|
de Oliveira JAGT, Neves MD, Sampaio GFS, Constantino CJL, Nakagaki WR. Analysis of the effects of turmeric and symbiotic consumption on bones of rats submitted to the use of dexamethasone. Life Sci 2022; 304:120690. [DOI: 10.1016/j.lfs.2022.120690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
|
5
|
Bone Cell Exosomes and Emerging Strategies in Bone Engineering. Biomedicines 2022; 10:biomedicines10040767. [PMID: 35453517 PMCID: PMC9033129 DOI: 10.3390/biomedicines10040767] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 01/27/2023] Open
Abstract
Bone tissue remodeling is a highly regulated process balancing bone formation and resorption through complex cellular crosstalk between resident bone and microenvironment cells. This cellular communication is mediated by direct cell and cell–matrix contact, autocrine, endocrine, and paracrine receptor mediated mechanisms such as local soluble signaling molecules and extracellular vesicles including nanometer sized exosomes. An impairment in this balanced process leads to development of pathological conditions. Bone tissue engineering is an emerging interdisciplinary field with potential to address bone defects and disorders by synthesizing three-dimensional bone substitutes embedded with cells for clinical implantation. However, current cell-based therapeutic approaches have faced hurdles due to safety and ethical concerns, challenging their clinical translation. Recent studies on exosome-regulated bone homeostasis and regeneration have gained interest as prospective cell free therapy in conjugation with tissue engineered bone grafts. However, exosome research is still in its nascent stages of bone tissue engineering. In this review, we specifically describe the role of exosomes secreted by cells within bone microenvironment such as osteoblasts, osteocytes, osteoclasts, mesenchymal stem cell cells, immune cells, endothelial cells, and even tumor cells during bone homeostasis and crosstalk. We also review exosome-based osteoinductive functionalization strategies for various bone-based biomaterials such as ceramics, polymers, and metals in bone tissue engineering. We further highlight biomaterials as carrier agents for exosome delivery to bone defect sites and, finally, the influence of various biomaterials in modulation of cell exosome secretome.
Collapse
|
6
|
Mestres G, Carter SSD, Hailer NP, Diez-Escudero A. A practical guide for evaluating the osteoimmunomodulatory properties of biomaterials. Acta Biomater 2021; 130:115-137. [PMID: 34087437 DOI: 10.1016/j.actbio.2021.05.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/29/2021] [Accepted: 05/20/2021] [Indexed: 12/17/2022]
Abstract
Biomaterials offer a promising approach to repair bone defects. Whereas traditional studies predominantly focused on optimizing the osteogenic capacity of biomaterials, less focus has been on the immune response elicited by them. However, the immune and skeletal systems extensively interact, a concept which is referred to as 'osteoimmunology'. This realization has fuelled the development of biomaterials with favourable osteoimmunomodulatory (OIM) properties, aiming to modulate the immune response and to support bone regeneration, thereby affecting the success of an implant. Given the plethora of in vitro assays used to evaluate the OIM properties of biomaterials, it may be challenging to select the right methods to produce conclusive results. In this review, we aim to provide a comprehensive and practical guide for researchers interested in studying the OIM properties of biomaterials in vitro. After a concise overview of the concept of osteoimmunology, emphasis is put on the methodologies that are regularly used to evaluate the OIM properties of biomaterials. First, a description of the most commonly used cell types and cell culture media is provided. Second, typical experimental set-ups and their relevant characteristics are discussed. Third, a detailed overview of the generally used methodologies and readouts, including cell type-specific markers and time points of analysis, is given. Finally, we highlight the promise of advanced approaches, namely microarrays, bioreactors and microfluidic-based systems, and the potential that these may offer to the osteoimmunology field. STATEMENT OF SIGNIFICANCE: Osteoimmunology focuses on the connection and communication between the skeletal and immune systems. This interaction has been recognized to play an important role in the clinical success of biomaterials, which has resulted in an increasing amount of research on the osteoimmunomodulatory (OIM) properties of biomaterials. However, the amount of literature makes it challenging to extract the information needed to design experiments from beginning to end, and to compare obtained results to existing work. This article intends to serve as a guide for those aiming to learn more about the commonly used experimental approaches in the field. We cover early-stage choices, such as cell types and experimental set-ups, but also discuss specific assays, including cell markers and time points of analysis.
Collapse
Affiliation(s)
- Gemma Mestres
- Division of Microsystems Technology, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden.
| | - Sarah-Sophia D Carter
- Division of Microsystems Technology, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden
| | - Nils P Hailer
- Ortholab, Department of Surgical Sciences-Orthopaedics, Uppsala University, 751 85 Uppsala, Sweden
| | - Anna Diez-Escudero
- Ortholab, Department of Surgical Sciences-Orthopaedics, Uppsala University, 751 85 Uppsala, Sweden
| |
Collapse
|
7
|
He M, Yang B, Huo F, Xie L, Yang M, Tian W. A novel coating with universal adhesion and inflammation-responsive drug release functions to manipulate the osteoimmunomodulation of implants. J Mater Chem B 2021; 9:5272-5283. [PMID: 34137425 DOI: 10.1039/d1tb00953b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The immune response elicited by the bone endoprosthesis is currently considered an important factor that affects its interfacial osteointegration. In this work, a metal-phenolic-based drug-loaded coating with universal adhesion properties and intelligent drug delivery feature was created to promote osteointegration by manipulating a beneficial osteoimmune microenvironment. A novel pro-drug with inflammation-responsive release function was firstly synthesized via the esterification reaction between tannic acid (TA) and indometacin (IND), and then the coating was developed by chelating it with Fe3+. In the normal biological environment, the coating was stable, while, in the inflammatory environment, the release of TA and IND motifs could be triggered by the overexpressed esterase. The released TA and IND displayed synergistic effects on macrophage polarization, leading to a downregulation expression of pro-inflammatory cytokines, and an upregulation expression of anti-inflammatory cytokines and osteogenic-related factors. When stimulated by a conditioned medium generated by macrophages seeded onto the coating, the osteogenic differentiation potential of BMSCs was significantly enhanced. Finally, the designed coating significantly promoted the osteointegration of the implant, demonstrated by the increase of the bone-implant contact by two times. Additionally, the coating was substrate-independent and can be formed within seconds without special equipment, thus, it showed great potential applications to endow advanced hard tissue implants with favorable osteoimmunomodulation.
Collapse
Affiliation(s)
- Min He
- State Key Laboratory of Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bo Yang
- State Key Laboratory of Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Fangjun Huo
- State Key Laboratory of Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li Xie
- State Key Laboratory of Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, P. R. China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China. and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
8
|
Tu Y, Yang Y, Li Y, He C. Naturally occurring coumestans from plants, their biological activities and therapeutic effects on human diseases. Pharmacol Res 2021; 169:105615. [PMID: 33872808 DOI: 10.1016/j.phrs.2021.105615] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/24/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023]
Abstract
Naturally occurring coumestans are known as a collection of plant-derived polycyclic aromatic secondary metabolites which are characterized by the presence of an oxygen heterocyclic four-ring system comprising a coumarin moiety and a benzofuran moiety sharing a C˭C bond. Recently, there is an increasing attention in excavating the medicinal potential of coumestans, particularly coumestrol, wedelolactone, psoralidin and glycyrol, in a variety of diseases. This review is a comprehensive inventory of the chemical structures of coumestans isolated from various plant sources during the period of 1956-2020, together with their reported biological activities. 120 molecules were collected and further classified as coumestans containing core skeleton, dimethylpyranocoumestans, furanocoumestans, O-glycosylated coumestans and others, which showed a wide range of pharmacological activities including estrogenic, anti-cancer, anti-inflammatory, anti-osteoporotic, organ protective, neuroprotective, anti-diabetic and anti-obesity, antimicrobial, immunosuppressive, antioxidant and skin-protective activities. Furthermore, this review focuses on the counteraction of coumestans against bone diseases and organ damages, and the involved molecular mechanisms, which could provide important information to better understand the medicinal values of these compounds. This review is intended to be instructive for the rational design and development of less toxic and more effective drugs with a coumestan scaffold.
Collapse
Affiliation(s)
- Yanbei Tu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Ying Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Yanfang Li
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR 999078, China.
| |
Collapse
|
9
|
Xie CL, Park KH, Kang SS, Cho KM, Lee DH. Isoflavone-enriched soybean leaves attenuate ovariectomy-induced osteoporosis in rats by anti-inflammatory activity. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:1499-1506. [PMID: 32851642 DOI: 10.1002/jsfa.10763] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/23/2020] [Accepted: 08/27/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND With an increasing aging population, postmenopausal osteoporosis has become a global public health problem. Previous evidence has shown that postmenopausal osteoporosis is a skeletal disease mainly caused by estrogen deficiency, generally accompanied by inflammation, and dietary isoflavones may ameliorate postmenopausal osteoporosis by anti-inflammatory activity. We have generated isoflavone-enriched soybean leaves (IESLs), but their anti-inflammatory activity and effect on attenuating osteoporosis are still obscure. Here, we determined the isoflavone profiles of IESLs and evaluated their anti-inflammatory activity in lipopolysaccharide-stimulated RAW 264.7 cells and anti-osteoporotic effects on ovariectomy-induced osteoporosis in rats. RESULTS IESLs had a high content of total isoflavone. Hydrolysate of IESLs (HIESLs) was rich with the aglycones daidzein and genistein, and HIESLs can significantly inhibit lipopolysaccharide-induced inflammation by reducing messenger RNA expression of iNOS, COX-2, IL6, and IL1β. Moreover, ovariectomized rats receiving aqueous extracts of IESLs (HIESLs) orally maintained more bone mass than control rats did, which was attributed to inhibition of osteoclastogenesis by downregulating the messenger RNA expression of the bone-specific genes RANKL/OPG, OC, and cathepsin K, and the inflammation-related genes IL6, NFκB, and COX-2. CONCLUSION IESLs may attenuate postmenopausal osteoporosis by suppressing osteoclastogenesis with anti-inflammatory activity and be a potential source of functional food ingredients for the prevention of osteoporosis. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Cheng-Liang Xie
- School of Pharmaceutical Science (Shenzhen), Sun Yat-sen University, Guangzhou, China
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Ki H Park
- Division of Applied Life Science (BK21 plus), IALS, Gyeongsang National University, Jinju, Republic of Korea
| | - Sang S Kang
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Kye M Cho
- Department of Food Science, Gyeongnam National University of Science and Technology, Jinju, Republic of Korea
| | - Dong H Lee
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
10
|
Epsley S, Tadros S, Farid A, Kargilis D, Mehta S, Rajapakse CS. The Effect of Inflammation on Bone. Front Physiol 2021; 11:511799. [PMID: 33584321 PMCID: PMC7874051 DOI: 10.3389/fphys.2020.511799] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
Bone remodeling is the continual process to renew the adult skeleton through the sequential action of osteoblasts and osteoclasts. Nuclear factor RANK, an osteoclast receptor, and its ligand RANKL, expressed on the surface of osteoblasts, result in coordinated control of bone remodeling. Inflammation, a feature of illness and injury, plays a distinct role in skewing this process toward resorption. It does so via the interaction of inflammatory mediators and their related peptides with osteoblasts and osteoclasts, as well as other immune cells, to alter the expression of RANK and RANKL. Such chemical mediators include TNFα, glucocorticoids, histamine, bradykinin, PGE2, systemic RANKL from immune cells, and interleukins 1 and 6. Conditions, such as periodontal disease and alveolar bone erosion, aseptic prosthetic loosening, rheumatoid arthritis, and some sports related injuries are characterized by the result of this process. A thorough understanding of bone response to injury and disease, and ability to detect such biomarkers, as well as imaging to identify early structural and mechanical property changes in bone architecture, is important in improving management and outcomes of bone related pathology. While gut health and vitamin and mineral availability appear vitally important, nutraceuticals also have an impact on bone health. To date most pharmaceutical intervention targets inflammatory cytokines, although strategies to favorably alter inflammation induced bone pathology are currently limited. Further research is required in this field to advance early detection and treatments.
Collapse
Affiliation(s)
- Scott Epsley
- Philadelphia 76ers, Philadelphia, PA, United States
| | - Samuel Tadros
- Department of Radiology and Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Alexander Farid
- Department of Radiology and Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Kargilis
- Department of Radiology and Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Chamith S. Rajapakse
- Department of Radiology and Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
11
|
Qian Z, Zhong Z, Ni S, Li D, Zhang F, Zhou Y, Kang Z, Qian J, Yu B. Cytisine attenuates bone loss of ovariectomy mouse by preventing RANKL-induced osteoclastogenesis. J Cell Mol Med 2020; 24:10112-10127. [PMID: 32790170 PMCID: PMC7520284 DOI: 10.1111/jcmm.15622] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/07/2020] [Accepted: 06/23/2020] [Indexed: 12/22/2022] Open
Abstract
Postmenopausal Osteoporosis (PMOP) is oestrogen withdrawal characterized of much production and activation by osteoclast in the elderly female. Cytisine is a quinolizidine alkaloid that comes from seeds or other plants of the Leguminosae (Fabaceae) family. Cytisine has been shown several potential pharmacological functions. However, its effects on PMOP remain unknown. This study designed to explore whether Cytisine is able to suppress RANKL-induced osteoclastogenesis and prevent the bone loss induced by oestrogen deficiency in ovariectomized (OVX) mice. In this study, we investigated the effect of Cytisine on RAW 264.7 cells and bone marrow monocytes (BMMs) derived osteoclast culture system in vitro and observed the effect of Cytisine on ovariectomized (OVX) mice model to imitate postmenopausal osteoporosis in vivo. We found that Cytisine inhibited F-actin ring formation and tartrate-resistant acid phosphatase (TRAP) staining in dose-dependent ways, as well as bone resorption by pit formation assays. For molecular mechanism, Cytisine suppressed RANK-related trigger RANKL by phosphorylation JNK/ERK/p38-MAPK, IκBα/p65-NF-κB, and PI3K/AKT axis and significantly inhibited these signalling pathways. However, the suppression of PI3K-AKT-NFATc1 axis was rescued by AKT activator SC79. Meanwhile, Cytisine inhibited RANKL-induced RANK-TRAF6 association and RANKL-related gene and protein markers such as NFATc1, Cathepsin K, MMP-9 and TRAP. Our study indicated that Cytisine could suppress bone loss in OVX mouse through inhibited osteoclastogenesis. All data provide the evidence that Cytisine may be a promising agent in the treatment of osteoclast-related diseases such as osteoporosis.
Collapse
Affiliation(s)
- Zhi Qian
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
- Department of Orthopaedic SurgeryZhangye People's Hospital affiliated to Hexi UniversityZhangye CityChina
| | - Zeyuan Zhong
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Shuo Ni
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Dejian Li
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Fangxue Zhang
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Ying Zhou
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Zhanrong Kang
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| | - Jun Qian
- Department of Orthopaedic SurgeryZhangye People's Hospital affiliated to Hexi UniversityZhangye CityChina
| | - Baoqing Yu
- Department of Orthopaedic SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterHuinan TownChina
| |
Collapse
|
12
|
Zhang W, Dang K, Huai Y, Qian A. Osteoimmunology: The Regulatory Roles of T Lymphocytes in Osteoporosis. Front Endocrinol (Lausanne) 2020; 11:465. [PMID: 32849268 PMCID: PMC7431602 DOI: 10.3389/fendo.2020.00465] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 06/15/2020] [Indexed: 12/23/2022] Open
Abstract
Immune imbalance caused bone loss. Osteoimmunology is emerging as a new interdisciplinary field to explore the shared molecules and interactions between the skeletal and immune systems. In particular, T lymphocytes (T cells) play pivotal roles in the regulation of bone health. However, the roles and mechanisms of T cells in the treatment of osteoporosis are not fully understood. The present review aims to summarize the essential regulatory roles of T cells in the pathophysiology of various cases of osteoporosis and the development of T cell therapy for osteoporosis from osteoimmunology perspective. As T cell-mediated immunomodulation inhibition reduced bone loss, there is an increasing interest in T cell therapy in an attempt to treat osteoporosis. In summary, the T cell therapy may be further pursued as an immunomodulatory strategy for the treatment of osteoporosis, which can provide a novel perspective for drug development in the future.
Collapse
Affiliation(s)
- Wenjuan Zhang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Kai Dang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Ying Huai
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Airong Qian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
13
|
Bouchet M, Lainé A, Boyault C, Proponnet-Guerault M, Meugnier E, Bouazza L, Kan CWS, Geraci S, El-Moghrabi S, Hernandez-Vargas H, Benetollo C, Yoshiko Y, Duterque-Coquillaud M, Clézardin P, Marie JC, Bonnelye E. ERRα Expression in Bone Metastases Leads to an Exacerbated Antitumor Immune Response. Cancer Res 2020; 80:2914-2926. [PMID: 32366476 DOI: 10.1158/0008-5472.can-19-3584] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/20/2020] [Accepted: 04/28/2020] [Indexed: 11/16/2022]
Abstract
Bone is the most common metastatic site for breast cancer. Although the estrogen-related receptor alpha (ERRα) has been implicated in breast cancer cell dissemination to the bone from the primary tumor, its role after tumor cell anchorage in the bone microenvironment remains elusive. Here, we reveal that ERRα inhibits the progression of bone metastases of breast cancer cells by increasing the immune activity of the bone microenvironment. Overexpression of ERRα in breast cancer bone metastases induced expression of chemokines CCL17 and CCL20 and repressed production of TGFβ3. Subsequently, CD8+ T lymphocytes recruited to bone metastases escaped TGFβ signaling control and were endowed with exacerbated cytotoxic features, resulting in significant reduction in metastases. The clinical relevance of our findings in mice was confirmed in over 240 patients with breast cancer. Thus, this study reveals that ERRα regulates immune properties in the bone microenvironment that contributes to decreasing metastatic growth. SIGNIFICANCE: This study places ERRα at the interplay between the immune response and bone metastases of breast cancer, highlighting a potential target for intervention in advanced disease.
Collapse
Affiliation(s)
- Mathilde Bouchet
- INSERM-UMR1033, Labex DEVweCAN, Lyon, France
- University of Lyon-France
| | - Alexandra Lainé
- University of Lyon-France
- Tumor Escape Resistance and Immunity Department, CRCL, INSERM 1052 CNRS 5286, Centre Léon Bérard, Labex DEVweCAN, Lyon, France
| | - Cyril Boyault
- Institute for Advanced Biosciences, UMR5209-INSERM1302, La Tronche, France
| | | | | | - Lamia Bouazza
- INSERM-UMR1033, Labex DEVweCAN, Lyon, France
- University of Lyon-France
| | - Casina W S Kan
- INSERM-UMR1033, Labex DEVweCAN, Lyon, France
- University of Lyon-France
| | - Sandra Geraci
- INSERM-UMR1033, Labex DEVweCAN, Lyon, France
- University of Lyon-France
| | | | - Hector Hernandez-Vargas
- Tumor Escape Resistance and Immunity Department, CRCL, INSERM 1052 CNRS 5286, Centre Léon Bérard, Labex DEVweCAN, Lyon, France
| | - Claire Benetollo
- University of Lyon-France
- INSERM-UMR5292 INSERM U1028, Lyon, France
| | - Yuji Yoshiko
- Department of Calcified Tissue Biology, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Japan
| | | | | | - Julien C Marie
- University of Lyon-France.
- Tumor Escape Resistance and Immunity Department, CRCL, INSERM 1052 CNRS 5286, Centre Léon Bérard, Labex DEVweCAN, Lyon, France
| | - Edith Bonnelye
- INSERM-UMR1033, Labex DEVweCAN, Lyon, France.
- University of Lyon-France
| |
Collapse
|
14
|
Millen AE, Pavlesen S. Could Vitamin D influence risk for Periodontal Disease - to "D" or not to "D"? CURRENT ORAL HEALTH REPORTS 2020; 7:98-111. [PMID: 33457183 PMCID: PMC7810034 DOI: 10.1007/s40496-020-00253-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss the literature on associations between vitamin D and periodontal disease, including its strengths and weaknesses. Future direction for continued work in this area is provided. RECENT FINDINGS Research in cross-sectional cohorts, surveys, and case-control studies provide support for a role of vitamin D in periodontal disease, especially using clinical indicators such as bleeding on probing and clinical attachment loss. However, these studies have a number of limitations. They cannot establish temporality of these associations. Most case-control studies have been limited in sample size and have inconsistent findings. A number of cross-sectional studies are restricted to select populations (e.g., persons with HIV, diabetes, rheumatoid arthritis) limiting extrapolation of findings to the general aging population.Fewer prospective studies have been conducted, and only three have examined associations using a biomarker for vitamin D that reflects exposure from all its sources (sunlight, diet and supplements). One study is limited by using self-reported measures of disease outcomes, and only two used measures of alveolar crestal height. However, of the prospective studies published, there is a suggestion that vitamin D might prevent against tooth loss. Only two randomized controlled trials have examined these associations, and they support the effects of vitamin D supplementation on prevention of tooth loss and gingival bleeding. SUMMARY We strongly suggest that new research should focus on prospective study designs with followup of participants longer than a decade and long-term clinical trials. Such studies should incorporate measures of alveolar bone loss and tooth loss with indication for reason for tooth loss. Such clinical trials should be designed to examine both the influence of vitamin D supplementation alone as well as with other nutrients (e.g., calcium) or therapeutic medications (e.g., bisphosphonates). Currently, there is no strong evidence to suggest that vitamin D protects against development of periodontal disease.
Collapse
Affiliation(s)
- Amy E Millen
- Farber Hall 270F, Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, NY. 14214
| | - Sonja Pavlesen
- Buffalo General Medical Center 6D501, 100 High St., Department of Orthopedic Surgery, Jacobs School of Medicine and Biomedical Science, University at Buffalo, State University of New York, Buffalo, NY. 14203
| |
Collapse
|
15
|
Gu H, Huang Z, Chen G, Zhou K, Zhang Y, Chen J, Xu J, Yin X. Network and pathway-based analyses of genes associated with osteoporosis. Medicine (Baltimore) 2020; 99:e19120. [PMID: 32080087 PMCID: PMC7034680 DOI: 10.1097/md.0000000000019120] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis (OP) is a disease characterized by bone mass loss, bone microstructure damage, increased bone fragility, and easy fracture. The molecular mechanism underlying OP remains unclear.In this study, we identified 217 genes associated with OP, and formed a gene set [OP-related genes gene set (OPgset)].The highly enriched GOs and pathways showed OPgset genes were significantly involved in multiple biological processes (skeletal system development, ossification, and osteoblast differentiation), and several OP-related pathways (Wnt signaling pathway, osteoclast differentiation, steroid hormone biosynthesis, and adipocytokine signaling pathway). Besides, pathway crosstalk analysis indicated three major modules, with first module consisted of pathways mainly involved in bone development-related signaling pathways, second module in Wnt-related signaling pathway and third module in metabolic pathways. Further, we calculated degree centrality of a node and selected ten key genes/proteins, including TGFB1, IL6, WNT3A, TNF, PTH, TP53, WNT1, IGF1, IL10, and SERPINE1. We analyze the K-core and construct three k-core sub-networks of OPgset genes.In summary, we for the first time explored the molecular mechanism underlying OP via network- and pathway-based methods, results from our study will improve our understanding of the pathogenesis of OP. In addition, these methods performed in this study can be used to explore pathogenesis and genes related to a specific disease.
Collapse
|
16
|
Ria R, Vacca A. Bone Marrow Stromal Cells-Induced Drug Resistance in Multiple Myeloma. Int J Mol Sci 2020; 21:ijms21020613. [PMID: 31963513 PMCID: PMC7013615 DOI: 10.3390/ijms21020613] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/12/2020] [Accepted: 01/14/2020] [Indexed: 01/06/2023] Open
Abstract
Multiple myeloma is a B-cell lineage cancer in which neoplastic plasma cells expand in the bone marrow and pathophysiological interactions with components of microenvironment influence many biological aspects of the malignant phenotype, including apoptosis, survival, proliferation, and invasion. Despite the therapeutic progress achieved in the last two decades with the introduction of a more effective and safe new class of drugs (i.e., immunomodulators, proteasome inhibitors, monoclonal antibodies), there is improvement in patient survival, and multiple myeloma (MM) remains a non-curable disease. The bone marrow microenvironment is a complex structure composed of cells, extracellular matrix (ECM) proteins, and cytokines, in which tumor plasma cells home and expand. The role of the bone marrow (BM) microenvironment is fundamental during MM disease progression because modification induced by tumor plasma cells is crucial for composing a "permissive" environment that supports MM plasma cells proliferation, migration, survival, and drug resistance. The "activated phenotype" of the microenvironment of multiple myeloma is functional to plasma cell proliferation and spreading and to plasma cell drug resistance. Plasma cell drug resistance induced by bone marrow stromal cells is mediated by stress-managing pathways, autophagy, transcriptional rewiring, and non-coding RNAs dysregulation. These processes represent novel targets for the ever-increasing anti-MM therapeutic armamentarium.
Collapse
Affiliation(s)
- Roberto Ria
- Correspondence: ; Tel.: +39-080-559-31-06; Fax: +39-080-559-38-04
| | | |
Collapse
|
17
|
Brunetti G, D'Amelio P, Mori G, Faienza MF. Editorial: Updates on Osteoimmunology: What's New on the Crosstalk Between Bone and Immune Cells. Front Endocrinol (Lausanne) 2020; 11:74. [PMID: 32153510 PMCID: PMC7045046 DOI: 10.3389/fendo.2020.00074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/03/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Giacomina Brunetti
- Section of Human Anatomy and Histology, Department of Basic and Medical Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Italy
- *Correspondence: Giacomina Brunetti
| | - Patrizia D'Amelio
- Gerontology Section, Department of Medical Sciences, University of Torino, Turin, Italy
- Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria Felicia Faienza
- Paediatric Unit, Department of Biomedical Science and Human Oncology, University of Bari, Bari, Italy
| |
Collapse
|
18
|
Huai Y, Zhang W, Chen Z, Zhao F, Wang W, Dang K, Xue K, Gao Y, Jiang S, Miao Z, Li M, Hao Q, Chen C, Qian A. A Comprehensive Analysis of MicroRNAs in Human Osteoporosis. Front Endocrinol (Lausanne) 2020; 11:516213. [PMID: 33193074 PMCID: PMC7609919 DOI: 10.3389/fendo.2020.516213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 09/14/2020] [Indexed: 01/09/2023] Open
Abstract
MicroRNAs (miRNAs) are single-stranded RNA molecules that control gene expression in various processes, such as cancers, Alzheimer's disease, and bone metabolic diseases. However, the regulatory roles of miRNAs in osteoporosis have not been systematically analyzed. Here, we performed a comprehensive analysis to identify the differentially expressed miRNAs involved in osteoporosis. MiRNAs associated with osteoporosis were collected through literature retrieval and further screened based on specific inclusion and exclusion criteria. The osteoporosis therapeutic targets of miRNAs were obtained by the integration of miRWalk 3.0 database and five human disease therapeutic target databases. Then, the network analysis and functional enrichment analysis of miRNAs and their targets were performed. As a result, 11 eligible miRNAs were identified highly associated with osteoporosis. MiRNA-mRNA network demonstrated there were the complex mutual interactions between miRNAs and their targets. Besides, ADRB2, AR, ESR1, FGFR1, TRAF6, etc., were identified as the top hub genes in protein-protein interaction (PPI) network. Functional enrichment analysis revealed that miRNAs and their targets were mainly mapped on processes associated with bone and immune system, such as bone remolding, bone mineralization, PI3K/AKt, TNF signaling pathways and Th17 cell differentiation. RT-PCR results showed that the expression of miR-335-3p was significantly down-regulated in hind limb unloading (HLU) mice tibia samples compared with controls, the remaining 10 miRNAs were significantly up-regulated after HLU (P < 0.01). In summary, we identified 11 differentially expressed miRNAs and their hub target genes in osteoporosis, which may be novel diagnostic biomarkers for osteoporosis.
Collapse
Affiliation(s)
- Ying Huai
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Wenjuan Zhang
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Zhihao Chen
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Fan Zhao
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Wei Wang
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Kai Dang
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Kaiyue Xue
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Yongguang Gao
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Shanfeng Jiang
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Zhiping Miao
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Chu Chen
- Clinical Laboratory of Honghui Hospital, Xi’an JiaoTong University College of Medicine, Xi’an, China
- *Correspondence: Airong Qian, ; Chu Chen,
| | - Airong Qian
- Lab for Bone Metabolism, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Airong Qian, ; Chu Chen,
| |
Collapse
|
19
|
Di Lullo G, Marcatti M, Heltai S, Tresoldi C, Paganoni AM, Bordignon C, Ciceri F, Protti MP. Immunomodulatory Drugs in the Context of Autologous Hematopoietic Stem Cell Transplantation Associate With Reduced Pro-tumor T Cell Subsets in Multiple Myeloma. Front Immunol 2019; 9:3171. [PMID: 30719025 PMCID: PMC6348257 DOI: 10.3389/fimmu.2018.03171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/24/2018] [Indexed: 01/07/2023] Open
Abstract
Immunomodulatory drugs (IMiDs) are effective therapeutics for multiple myeloma (MM), where in different clinical settings they exert their function both directly on MM cells and indirectly by modulating immune cell subsets, although with not completely defined mechanisms. Here we studied the role of IMiDs in the context of autologous hematopoietic stem cell transplantation on the T cell subset distribution in the bone marrow of newly diagnosed MM patients. We found that after transplantation pro-tumor Th17-Th1 and Th22 cells and their related cytokines were lower in patients treated with IMiDs during induction chemotherapy compared to untreated patients. Of note, lower levels of IL-17, IL-22, and related IL-6, TNF-α, IL-1β, and IL-23 in the bone marrow sera correlated with treatment with IMiDs and favorable clinical outcome. Collectively, our results suggest a novel anti-inflammatory role for IMiDs in MM.
Collapse
Affiliation(s)
- Giulia Di Lullo
- Tumor Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Magda Marcatti
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Heltai
- Tumor Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Tresoldi
- Molecular Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anna Maria Paganoni
- Laboratory for Modeling and Scientific Computing (MOX), Dipartimento di Matematica,Politecnico di Milano, Milan, Italy
| | | | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Maria Pia Protti
- Tumor Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
20
|
Han Y, Jin Y, Miao Y, Shi T, Lin X. Improved RANKL production by memory B cells: A way for B cells promote alveolar bone destruction during periodontitis. Int Immunopharmacol 2018; 64:232-237. [DOI: 10.1016/j.intimp.2018.08.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 08/02/2018] [Accepted: 08/23/2018] [Indexed: 12/20/2022]
|
21
|
Khera A, Kanta P, Kalra J, Dumir D, M T. Resveratrol restores the level of key inflammatory cytokines and RANKL/OPG ratio in the femur of rat osteoporosis model. J Women Aging 2018; 31:540-552. [PMID: 30239309 DOI: 10.1080/08952841.2018.1522126] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Resveratrol's effect on bone mineral density (BMD) and expression of cytokines in ovariectomized rats (postmenopausal osteoporosis model) was studied. The study was conducted on 3-month-old Sprague-Dawley rats that were (a) sham-operated, (b) ovariectomized, (c) ovariectomized and treated with β-estradiol (487.5 µg/kg weight/day), and (d) ovariectomized and treated with resveratrol (625 µg/Kg body weight/day). The treatment was for 4 weeks. After sacrifice BMD and gene expression (RANKL, OPG, IL-23, and IL-17A, IL-1β, and TNFα) were measured in tibia and femur respectively. Resveratrol could restore RANKL/OPG ratio, slightly increase BMD, and moderately but significantly reduce IL-23, IL-17A, IL-1β, and TNF-α cytokine expression levels.
Collapse
Affiliation(s)
- Alka Khera
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Poonam Kanta
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jaswinder Kalra
- Department of Obstetrics and Gynecology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Dheeraj Dumir
- Max Super Specialty Hospital, Max Institute of Cardiology, Mohali, Punjab, India
| | - Thungapathra M
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
22
|
Chen K, Lv ZT, Cheng P, Zhu WT, Liang S, Yang Q, Parkman VJA, Zhou CH, Jing XZ, Liu H, Wang YT, Lin H, Liao H, Chen AM. Boldine Ameliorates Estrogen Deficiency-Induced Bone Loss via Inhibiting Bone Resorption. Front Pharmacol 2018; 9:1046. [PMID: 30271347 PMCID: PMC6146032 DOI: 10.3389/fphar.2018.01046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 08/27/2018] [Indexed: 12/31/2022] Open
Abstract
Osteoporosis is an enormous health problem caused by the imbalance between bone resorption and bone formation. The current therapeutic strategies for osteoporosis still have some limitations. Boldine, an alkaloid isolated from Peumus boldus, has been shown to have antioxidant and anti-inflammatory effects in vivo. For the first time, we discover that boldine has a protective effect for the estrogen deficiency-induced bone loss in mice. According to the Micro-CT and histomorphometry assays, boldine conducts this protective effect through inhibiting bone resorption without affecting bone formation in vivo. Moreover, we showed that boldine can inhibit receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation via impairing the AKT signaling pathways, while SC79 (an AKT agonist) partially rescue this effect. In conclusion, our results suggest that boldine can prevent estrogen deficiency-induced osteoporosis by inhibiting osteoclastogenesis. Thus, boldine may be served as a novel therapeutic agent for anti-osteoporotic therapy.
Collapse
Affiliation(s)
- Kun Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Engineering and Regenerative Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, United States
| | - Zheng-tao Lv
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Engineering and Regenerative Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-tao Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Liang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Engineering and Regenerative Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Virginia-Jeni Akila Parkman
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, United States
| | - Chen-he Zhou
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xing-zhi Jing
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Engineering and Regenerative Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-ting Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Engineering and Regenerative Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Lin
- Department of Orthopaedic Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Liao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - An-min Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Biological Engineering and Regenerative Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Jiang WY, Xing C, Wang HW, Wang W, Chen SZ, Ning LF, Xu X, Tang QQ, Huang HY. A Lox/CHOP-10 crosstalk governs osteogenic and adipogenic cell fate by MSCs. J Cell Mol Med 2018; 22:5097-5108. [PMID: 30044535 PMCID: PMC6156357 DOI: 10.1111/jcmm.13798] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 05/31/2018] [Accepted: 06/23/2018] [Indexed: 12/20/2022] Open
Abstract
Accelerated marrow adipogenesis has been associated with ageing and osteoporosis and is thought to be because of an imbalance between adipogenic and osteogenic differentiation of mesenchymal stem cell (MSCs). We have previously found that lysyl oxidase (Lox) inhibition disrupts BMP4‐induced adipocytic lineage commitment and differentiation of MSCs. In this study, we found that lox inhibition dramatically up‐regulates BMP4‐induced expression of CCAAT/enhancer binding protein (C/EBP) homologous protein 10 (CHOP‐10), which then promotes BMP4‐induced osteogenesis of MSCs both in vitro and in vivo. Specifically, Lox inhibition or CHOP‐10 up‐regulation activated Wnt/β‐catenin signalling to enhance BMP4‐induced osteogenesis, with pro‐adipogenic p38 MAPK and Smad signalling suppressed. Together, we demonstrate that Lox/CHOP‐10 crosstalk regulates BMP4‐induced osteogenic and adipogenic fate determination of MSCs, presenting a promising therapeutic target for osteoporosis and other bone diseases.
Collapse
Affiliation(s)
- Wen-Yan Jiang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Shanghai, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chun Xing
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Shanghai, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hong-Wei Wang
- Biliary and Pancreatic Center, Huadong Hospital, Fudan University, Shanghai, China
| | - Wei Wang
- Biliary and Pancreatic Center, Huadong Hospital, Fudan University, Shanghai, China
| | - Su-Zhen Chen
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Shanghai, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Liu-Fang Ning
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Shanghai, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xu Xu
- Institute of Stem Cell Research and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Shanghai, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Institute of Stem Cell Research and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hai-Yan Huang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education, Shanghai, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Institute of Stem Cell Research and Regenerative Medicine, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Abstract
The gut microbiota (GM) is the whole of commensal, symbiotic, and pathogenic microorganisms living in our intestine. The GM-host interactions contribute to the maturation of the host immune system, modulating its systemic response. It is well documented that GM can interact with non-enteral cells such as immune cells, dendritic cells, and hepatocytes, producing molecules such as short-chain fatty acids, indole derivatives, polyamines, and secondary bile acid. The receptors for some of these molecules are expressed on immune cells, and modulate the differentiation of T effector and regulatory cells: this is the reason why dysbiosis is correlated with several autoimmune, metabolic, and neurodegenerative diseases. Due to the close interplay between immune and bone cells, GM has a central role in maintaining bone health and influences bone turnover and density. GM can improve bone health also increasing calcium absorption and modulating the production of gut serotonin, a molecule that interacts with bone cells and has been suggested to act as a bone mass regulator. Thus, GM manipulation by consumption of antibiotics, changes in dietary habits, and the use of pre- and probiotics may affect bone health. This review summarizes evidences on the influence of GM on immune system and on bone turnover and density and how GM manipulation may influence bone health.
Collapse
Affiliation(s)
- P D'Amelio
- Gerontology and Bone Metabolic Diseases Section, Department of Medical Science, University of Torino, CorsoDogliotti 14, 10126, Turin, Italy.
| | - F Sassi
- Gerontology and Bone Metabolic Diseases Section, Department of Medical Science, University of Torino, CorsoDogliotti 14, 10126, Turin, Italy
| |
Collapse
|
25
|
Wang J, Wu X, Duan Y. Magnesium Lithospermate B Protects against Lipopolysaccharide-Induced Bone Loss by Inhibiting RANKL/RANK Pathway. Front Pharmacol 2018; 9:64. [PMID: 29472860 PMCID: PMC5810254 DOI: 10.3389/fphar.2018.00064] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/17/2018] [Indexed: 12/12/2022] Open
Abstract
Lipopolysaccharide (LPS) can induce bone loss by stimulating bone resorption. Natural compounds have great potential for the treatment of osteolytic bone diseases. Magnesium lithospermate B (MLB) plays an important role in protecting against oxidative damage and also has potential anti-inflammatory pharmacological properties. However, its role in LPS-induced bone loss is still unknown. In the present study, we observed the effects of MLB on LPS-induced bone damage and investigated the possible mechanisms. The bone loss models were established by LPS administration in male Sprague–Dawley rats. MLB (200 mg/kg body weight) was given by subcutaneous injection. MicroCT analysis, biomarker assay, histological examination and immunohistochemical staining were performed at the 8th weeks. In addition, RAW264.7 cells were treated with LPS in the presence or absence of MLB. The osteoclast formation, resorption activity and differentiation-related genes [(receptor activator of nuclear factor kappa-B (RANK), Traf6, Fra-1, and c-src)] expression were evaluated. LPS induced bone loss shown as the decrease in bone volume fraction and trabecular number, and increase in trabecular separation. LPS also markedly enhanced the osteoclast formation and resorption activity compared with the control. MLB significantly abolished the LPS-induced bone microstructure damage (p < 0.05) and osteoclast formation. MLB also inhibited the increases of serum tartrate-resistant acid phosphatase 5b, RANK ligand (RANKL) and TNF-α level enhanced by LPS (p < 0.05). Immunohistochemical staining indicated that MLB attenuated the high expression of RANKL and RANK stimulated by LPS. In addition, MLB significantly abolished the LPS-enhanced osteoclast formation, resorption activity, RANK, Traf6, Fra-1, and c-src expression in vitro. Our data demonstrate that MLB can suppress LPS-induced bone loss via inhibiting RANKL/RANK related osteoclast formation.
Collapse
Affiliation(s)
- Jihai Wang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhenghou, China
| | - Xuejian Wu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhenghou, China
| | - Yongzhuang Duan
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhenghou, China
| |
Collapse
|
26
|
Abstract
Breast cancer bone metastasis develops as the result of a series of complex interactions between tumor cells, bone marrow cells, and resident bone cells. The net effect of these interactions are the disruption of normal bone homeostasis, often with significantly increased osteoclast and osteoblast activity, which has provided a rational target for controlling tumor progression, with little or no emphasis on tumor eradication. Indeed, the clinical course of metastatic breast cancer is relatively long, with patients likely to experience sequential skeletal-related events (SREs), often over lengthy periods of time, even up to decades. These SREs include bone pain, fractures, and spinal cord compression, all of which may profoundly impair a patient's quality-of-life. Our understanding of the contributions of the host bone and bone marrow cells to the control of tumor progression has grown over the years, yet the focus of virtually all available treatments remains on the control of resident bone cells, primarily osteoclasts. In this perspective, our focus is to move away from the current emphasis on the control of bone cells and focus our attention on the hallmarks of bone metastatic tumor cells and how these differ from primary tumor cells and normal host cells. In our opinion, there remains a largely unmet medical need to develop and utilize therapies that impede metastatic tumor cells while sparing normal host bone and bone marrow cells. This perspective examines the impact of metastatic tumor cells on the bone microenvironment and proposes potential new directions for uncovering the important mechanisms driving metastatic progression in bone based on the hallmarks of bone metastasis.
Collapse
Affiliation(s)
- Rachelle W Johnson
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Larry J Suva
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
27
|
Fessler J, Husic R, Schwetz V, Lerchbaum E, Aberer F, Fasching P, Ficjan A, Obermayer-Pietsch B, Duftner C, Graninger W, Stradner MH, Dejaco C. Senescent T-Cells Promote Bone Loss in Rheumatoid Arthritis. Front Immunol 2018; 9:95. [PMID: 29472917 PMCID: PMC5810289 DOI: 10.3389/fimmu.2018.00095] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/12/2018] [Indexed: 12/18/2022] Open
Abstract
Objective T-cells are critical players in the pathogenesis of osteoporosis in patients with rheumatoid arthritis (RA). Premature senescence of lymphocytes including the accumulation of senescent CD4+ T-cells is a hallmark feature of RA. Whether T-cell senescence is associated with bone loss in RA patients is elusive so far. Methods This includes a prospective study of consecutive patients with RA (n = 107), patients with primary osteopenia/-porosis (n = 75), and healthy individuals (n = 38). Bone mineral density (BMD) was determined by dual-energy X-ray absorptiometry scan. Flow cytometry, magnetic-associated cell sorting, and cell culture experiments were performed to analyze the pro-osteoclastic phenotype and the function of senescent CD4+CD28− T-cells. Results Patients with osteopenia/-porosis yielded a higher prevalence of senescent CD4+CD28− T-cells than individuals with normal BMD, in the RA, as well as in the non-RA cohort. Receptor activator of nuclear factor kappa-B ligand (RANKL) was expressed at higher levels on CD4+CD28− T-cells as compared to CD28+ T-cells. Stimulation with interleukin-15 led to an up-regulation of RANKL expression, particularly on CD28− T-cells. CD4+CD28− T-cells induced osteoclastogenesis more efficiently than CD28+ T-cells. Conclusion Our data indicate that senescent T-cells promote osteoclastogenesis more efficiently than conventional CD28+ T-cells, which might contribute to the pathogenesis of systemic bone loss in RA and primary osteoporosis.
Collapse
Affiliation(s)
- Johannes Fessler
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Rusmir Husic
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Verena Schwetz
- Department of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Elisabeth Lerchbaum
- Department of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Felix Aberer
- Department of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Patrizia Fasching
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Anja Ficjan
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | | | - Christina Duftner
- Department of Internal Medicine VI, Innsbruck Medical University, Innsbruck, Austria
| | - Winfried Graninger
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | | | - Christian Dejaco
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria.,Rheumatology Service, South Tyrolean Health Trust, Hospital Bruneck, Bruneck, Italy
| |
Collapse
|
28
|
Zhao L, Cai C, Wang J, Zhao L, Li W, Liu C, Guan H, Zhu Y, Xiao J. Dihydromyricetin Protects against Bone Loss in Ovariectomized Mice by Suppressing Osteoclast Activity. Front Pharmacol 2017; 8:928. [PMID: 29311931 PMCID: PMC5742133 DOI: 10.3389/fphar.2017.00928] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/07/2017] [Indexed: 12/21/2022] Open
Abstract
Dihydromyricetin (DMY), the main flavonoid component of Ampelopsis grossedentata, possesses pharmacological activities useful for treatment of diseases associated with inflammation and oxidative damage. Because osteoclasts are often involved in chronic low-grade systemic inflammation and oxidative damage, we hypothesized that DMY may be an effective treatment for osteoclast-related diseases. The effects of DMY on osteoclast formation and activity were examined in vitro. Female C57BL/6 mice were ovariectomized to mimic menopause-induced bone loss and treated with DMY, and femur samples were subjected to bone structure and histological analysis, serum biochemical indicators were also measured. DMY suppressed the activation of nuclear factor-κB, c-Fos and mitogen-activated protein kinase, and prevented production of reactive oxygen species. DMY decreased expression of osteoclast-specific genes, including Trap, Mmp-9, Cathepsin K, C-Fos, Nfatc1, and Rank. In addition, DMY prevented bone loss and decreased serum levels of tumor necrosis factor-α, interleukin-1β, and interleukin-6, and with a decrease in the ratio between receptor activator of nuclear factor-κB (RANK) ligand (RANKL) and osteoprotegerin (OPG) in vivo. These findings demonstrate that DMY attenuates bone loss and inhibits osteoclast formation and activity through modulation of multiple pathways both upstream and downstream of RANKL signaling. DMY may thus be a useful option for treatment of osteoclast-related diseases such as rheumatoid arthritis and osteoporosis.
Collapse
Affiliation(s)
- Libo Zhao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Cai
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of Oncology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Liming Zhao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijin Li
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changyu Liu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hanfeng Guan
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanli Zhu
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Xiao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Brunetti G, Faienza MF, Colaianni G, Grano M, Colucci S. Mechanisms of Altered Bone Remodeling in Multiple Myeloma. Clin Rev Bone Miner Metab 2017. [DOI: 10.1007/s12018-017-9236-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
30
|
Horváthová M, Ilavská S, Štefíková K, Szabová M, Krivošíková Z, Jahnová E, Tulinská J, Spustová V, Gajdoš M. The Cell Surface Markers Expression in Postmenopausal Women and Relation to Obesity and Bone Status. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14070751. [PMID: 28696349 PMCID: PMC5551189 DOI: 10.3390/ijerph14070751] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 06/30/2017] [Accepted: 06/30/2017] [Indexed: 12/16/2022]
Abstract
The age-related changes and hormonal deprivation in postmenopausal women are associated with the immune response alteration. The excessive fat accumulation, local and systemic inflammation may lead to dysregulation in immune function and relevant health problems, including obesity and osteoporosis. We analyzed the expression of cell surface markers in the venous blood specimens, stained with fluorophores-conjugated monoclonal antibodies and analysed by multicolour flow cytometry. The significant changes of cytotoxic, naive, and memory T-lymphocytes, plasmacytoid dendritic cells (DCs) were in postmenopausal women versus fertile women. Body mass index (BMI) affected markedly the cell surface expression of CD265/RANK. Osteoporosis is linked to reduced percentage of plasmacytoid DCs, and elevated natural Treg cells (p < 0.05). The confounding factors such as women age, BMI, bone mineral density (BMD), waist size and tissue fat affect the expression of RANK on myeloid DCs and CD40L on T-lymphocytes that might be the immunophenotypic modulators after menopause.
Collapse
Affiliation(s)
- Mira Horváthová
- Department of Immunology and Immunotoxicology, Faculty of Medicine, Slovak Medical University, 833 03 Bratislava, Slovakia.
| | - Silvia Ilavská
- Department of Immunology and Immunotoxicology, Faculty of Medicine, Slovak Medical University, 833 03 Bratislava, Slovakia.
| | - Kornélia Štefíková
- Department of Clinical and Experimental Pharmacology, Faculty of Medicine, Slovak Medical University, 83303 Bratislava, Slovakia.
| | - Michaela Szabová
- Department of Immunology and Immunotoxicology, Faculty of Medicine, Slovak Medical University, 833 03 Bratislava, Slovakia.
| | - Zora Krivošíková
- Department of Clinical and Experimental Pharmacology, Faculty of Medicine, Slovak Medical University, 83303 Bratislava, Slovakia.
| | - Eva Jahnová
- Department of Immunology and Immunotoxicology, Faculty of Medicine, Slovak Medical University, 833 03 Bratislava, Slovakia.
| | - Jana Tulinská
- Department of Immunology and Immunotoxicology, Faculty of Medicine, Slovak Medical University, 833 03 Bratislava, Slovakia.
| | - Viera Spustová
- Department of Clinical and Experimental Pharmacology, Faculty of Medicine, Slovak Medical University, 83303 Bratislava, Slovakia.
| | - Martin Gajdoš
- Department of Clinical and Experimental Pharmacology, Faculty of Medicine, Slovak Medical University, 83303 Bratislava, Slovakia.
| |
Collapse
|
31
|
Delion M, Braux J, Jourdain ML, Guillaume C, Bour C, Gangloff S, Pimpec-Barthes FL, Sermet-Gaudelus I, Jacquot J, Velard F. Overexpression of RANKL in osteoblasts: a possible mechanism of susceptibility to bone disease in cystic fibrosis. J Pathol 2017; 240:50-60. [PMID: 27235726 DOI: 10.1002/path.4753] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/09/2016] [Accepted: 05/18/2016] [Indexed: 12/19/2022]
Abstract
Bone fragility and loss are a significant cause of morbidity in patients with cystic fibrosis (CF), and the lack of effective therapeutic options means that treatment is more often palliative rather than curative. A deeper understanding of the pathogenesis of CF-related bone disease (CFBD) is necessary to develop new therapies. Defective CF transmembrane conductance regulator (CFTR) protein and chronic inflammation in bone are important components of the CFBD development. The receptor activator of nuclear factor kappa-B ligand (RANKL) and osteoprotegerin (OPG) drive the regulation of bone turnover. To investigate their roles in CFBD, we evaluated the involvement of defective CFTR in their production level in CF primary human osteoblasts with and without inflammatory stimulation, in the presence or not of pharmacological correctors of the CFTR. No major difference in cell ultrastructure was noted between cultured CF and non-CF osteoblasts, but a delayed bone matrix mineralization was observed in CF osteoblasts. Strikingly, resting CF osteoblasts exhibited strong production of RANKL protein, which was highly localized at the cell membrane and was enhanced in TNF (TNF-α) or IL-17-stimulated conditions. Under TNF stimulation, a defective response in OPG production was observed in CF osteoblasts in contrast to the elevated OPG production of non-CF osteoblasts, leading to an elevated RANKL-to-OPG protein ratio in CF osteoblasts. Pharmacological inhibition of CFTR chloride channel conductance in non-CF osteoblasts replicated both the decreased OPG production and the enhanced RANKL-to-OPG ratio. Interestingly, using CFTR correctors such as C18, we significantly reduced the production of RANKL by CF osteoblasts, in both resting and TNF-stimulated conditions. In conclusion, the overexpression of RANKL and high membranous RANKL localization in osteoblasts are related to defective CFTR, and may worsen bone resorption, leading to bone loss in patients with CF. Targeting osteoblasts with CFTR correctors may represent an effective strategy to treat CFBD. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Martial Delion
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Julien Braux
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Marie-Laure Jourdain
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Christine Guillaume
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Camille Bour
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Sophie Gangloff
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | | | - Isabelle Sermet-Gaudelus
- Unité de Pneumo-Pédiatrie Allergologie, Hôpital Necker, Inserm U1551, Université Paris Sorbonne, Paris, France
| | - Jacky Jacquot
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| | - Frédéric Velard
- EA 4691, Biomatériaux et Inflammation en Site Osseux, SFR CAP-Santé (FED 4231), Université Reims Champagne-Ardenne, 1 Avenue du Maréchal Juin, Reims, France
| |
Collapse
|
32
|
Kamycheva E, Goto T, Camargo CA. Celiac disease is associated with reduced bone mineral density and increased FRAX scores in the US National Health and Nutrition Examination Survey. Osteoporos Int 2017; 28:781-790. [PMID: 27714440 DOI: 10.1007/s00198-016-3791-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 09/25/2016] [Indexed: 12/17/2022]
Abstract
UNLABELLED We investigated the association between celiac disease (CD) and bone mass density (BMD) and risk of osteoporotic fractures in the general US population. In children and men ≥18 years, CD was associated with reduced BMD, and in men ≥40 years, CD was associated with increased risk of osteoporotic fractures. INTRODUCTION Celiac disease (CD) is an autoimmune condition, characterized by inflammation of the small intestine. CD has an increasing prevalence, and if unrecognized or untreated, CD can lead to complications from malabsorption and micronutrient deficiencies. We aimed to study whether CD is an independent predictor of reduced bone mineral density (BMD) and FRAX scores in the general US population. METHODS We used data from the National Health and Nutrition Examination Survey, 2009-2010 and 2013-2014. CD was defined by positive tissue transglutaminase IgA antibody test. Multivariable models of BMD and FRAX scores were adjusted for BMI, serum 25-hydroxyvitamin D, vitamin D and calcium supplements, milk intake, serum calcium, and smoking status, when available. RESULTS In children, aged 8-17 years, CD was associated with decreased Z-scores, by 0.85 for hip and 0.46 for spine (both P < 0.001). In men aged ≥ 18 years, CD was associated with 0.06 g/cm2 decrease in BMD in hip and with 0.11 g/cm2 decrease in BMD in spine (P = 0.08 and P < 0.001, respectively). In women, there were no statistically significant differences in the multiple-adjusted model. In men aged ≥ 40 years, CD predicted FRAX scores, resulting in increased scores by 2.25 % (P = 0.006) for hip fracture and by 2.43 % (P = 0.05) for major osteoporotic fracture. CD did not predict FRAX scores in women aged ≥40 years. CONCLUSION CD is independently associated with reduced BMD in children and adults aged ≥18 years and is an independent risk factor of osteoporotic fractures in men aged ≥40 years.
Collapse
Affiliation(s)
- E Kamycheva
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, 125 Nashua St, Suite 920, Boston, MA, 02114, USA.
- Medical Clinic, University Hospital of North Norway, Sykehusveien 38, 9038, Tromsoe, Norway.
- Endocrine Research Group, Faculty of Health Sciences, Department of Clinical Medicine, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9017, Tromsoe, Norway.
| | - T Goto
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, 125 Nashua St, Suite 920, Boston, MA, 02114, USA
- Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - C A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, 125 Nashua St, Suite 920, Boston, MA, 02114, USA
- Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| |
Collapse
|
33
|
|
34
|
Loss of stat3 function leads to spine malformation and immune disorder in zebrafish. Sci Bull (Beijing) 2017; 62:185-196. [PMID: 36659403 DOI: 10.1016/j.scib.2017.01.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 11/22/2016] [Accepted: 12/08/2016] [Indexed: 01/21/2023]
Abstract
STAT (Signal Transducers and Activators of Transcription) gene family members have been revealed to be involved in cell growth and differentiation in vertebrates. Despite their physiological importance, their functions are poorly studied at organ and systemic levels. In this study, we performed a genome-wide analysis using data from invertebrates to vertebrates to identify STAT genes and analyze their evolutionary history. Interestingly, the STAT gene family undergoes genome duplications during the evolutionary history with STAT3 homologues firstly appearing in the basal extant vertebrate, sea lamprey, suggesting its possible roles in spine formation. To investigate the functions of stat3 in fish species, TALEN technology was performed to generate mutant zebrafish lines. Stat3 mutant zebrafish showed no obvious defects at early developmental stage but displayed severe lateral and vertical curvature of the spine (scoliosis), spine fracture and the incomplete bone joints with narrower junction between vertebrae at early juvenile stage, as indicated by Alizarin red and Alcian blue staining, radiography and micro-computed tomography (MicroCT) analysis. Transcriptome analysis reveals dramatic alterations in a number of genes involved in immune and infection response, skeletal development and somatic growth, especially downregulated expression of collagen gene family, in the juvenile stat3 mutant zebrafish. Moreover, most of the collagen genes were detected to have abnormal expression pattern during the formation of spine deformities in stat3 mutants. Our data reveal that stat3 is specially expressed in vertebrates and required for normal spine development and immune function in zebrafish.
Collapse
|
35
|
Anitha D, Thomas B, Jan KS, Subburaj K. Risk of vertebral compression fractures in multiple myeloma patients: A finite-element study. Medicine (Baltimore) 2017; 96:e5825. [PMID: 28079810 PMCID: PMC5266172 DOI: 10.1097/md.0000000000005825] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The purpose of this study was to develop and validate a finite element (FE) model to predict vertebral bone strength in vitro using multidetector computed tomography (MDCT) images in multiple myeloma (MM) patients, to serve as a complementing tool to assess fracture risk. In addition, it also aims to differentiate MM patients with and without vertebral compression fractures (VCFs) by performing FE analysis on vertebra segments (T1-L5) obtained from in vivo routine MDCT imaging scans. MDCT-based FE models were developed from the in vitro vertebrae samples and were then applied to the in vivo vertebrae segments of MM patients (n = 4) after validation. Predicted fracture load using FE models correlated significantly with experimentally measured failure load (r = 0.85, P < 0.001). Interestingly, an erratic behavior was observed in patients with fractures (n = 2) and a more gradual change in FE-predicted strength values in patients without fractures (n = 2). Severe geometric deformations were also observed in models that have already attained fractures. Since BMD is not a reliable parameter for fracture risk prediction in MM subjects, it is necessary to use advanced tools such as FE analysis to predict individual fracture risk. If peaks are observed between adjacent segments in an MM patient, it can be safe to conclude that the spine is experiencing regions of structural instability. Such an FE visualization may have therapeutic consequences to prevent MM associated vertebral fractures.
Collapse
Affiliation(s)
- D. Anitha
- Engineering Product Development (EPD), Singapore University of Technology and Design (SUTD), Singapore
| | - Baum Thomas
- Department of Radiology, Klinikum rechts der Isar, Technische Universitaet Muenchen, Muenchen, Germany
| | - Kirschke S. Jan
- Department of Radiology, Klinikum rechts der Isar, Technische Universitaet Muenchen, Muenchen, Germany
| | - Karupppasamy Subburaj
- Engineering Product Development (EPD), Singapore University of Technology and Design (SUTD), Singapore
| |
Collapse
|
36
|
Brunetti G, D’Amelio P, Wasniewska M, Mori G, Faienza MF. Editorial: Bone: Endocrine Target and Organ. Front Endocrinol (Lausanne) 2017; 8:354. [PMID: 29312144 PMCID: PMC5742131 DOI: 10.3389/fendo.2017.00354] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 12/07/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Giacomina Brunetti
- Department of Basic and Medical Sciences, Neurosciences and Sense Organs, Section of Human Anatomy and Histology, University of Bari, Bari, Italy
- *Correspondence: Giacomina Brunetti,
| | - Patrizia D’Amelio
- Department of Medical Sciences, Gerontology Section, University of Torino, Torino, Italy
| | | | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria Felicia Faienza
- Department of Biomedical Science and Human Oncology, Paediatric Unit, University of Bari, Bari, Italy
| |
Collapse
|
37
|
Xu YY, Zeng QB, Yao ML, Yu C, Li J, Zhang AH. A possible new mechanism and drug intervention for kidney damage due to arsenic poisoning in rats. Toxicol Res (Camb) 2016; 5:511-518. [PMID: 30090365 PMCID: PMC6061977 DOI: 10.1039/c5tx00165j] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 12/30/2015] [Indexed: 11/21/2022] Open
Abstract
Arsenic poisoning is a worldwide endemic disease that affects thousands of people. Currently, the aetiology of the disease is known, but its pathogenesis is uncharacterized and there is no specific treatment. We established a rat model of coal-burning arsenic poisoning by feeding the animals corn powder baked with high arsenic coal. By observing subsequent changes in kidney and immune function, we found that arsenic induces both kidney and immune damage. Furthermore, there is a significant correlation between kidney and immune damage. Moreover, Ginkgo biloba, a known immune enhancer, was used as an intervention agent in arsenic poisoned rats to validate the relationship between kidney and immune damage. Meanwhile, we also explored the mechanism of Ginkgo biloba treatment of kidney damage in burning-coal arsenic poisoned rats. We found that Ginkgo biloba enhanced immune function in rats with arsenic poisoning and ameliorated arsenic-induced kidney damage. These results suggest that immune suppression may be one of the mechanisms underlying arsenic-induced kidney damage and that Ginkgo biloba might relieve kidney damage by enhancing immune function.
Collapse
Affiliation(s)
- Yu-Yan Xu
- Key Laboratory of Environment Pollution Monitoring and Disease Control , Ministry of Education; Department of Toxicology , School of Public Health , Guizhou Medical University , Guiyang , Guizhou 550025 , China . ; ; Tel: +86 851 8841 6172
| | - Qi-Bing Zeng
- Key Laboratory of Environment Pollution Monitoring and Disease Control , Ministry of Education; Department of Toxicology , School of Public Health , Guizhou Medical University , Guiyang , Guizhou 550025 , China . ; ; Tel: +86 851 8841 6172
| | - Mao-Lin Yao
- Key Laboratory of Environment Pollution Monitoring and Disease Control , Ministry of Education; Department of Toxicology , School of Public Health , Guizhou Medical University , Guiyang , Guizhou 550025 , China . ; ; Tel: +86 851 8841 6172
| | - Chun Yu
- Key Laboratory of Environment Pollution Monitoring and Disease Control , Ministry of Education; Department of Toxicology , School of Public Health , Guizhou Medical University , Guiyang , Guizhou 550025 , China . ; ; Tel: +86 851 8841 6172
| | - Jun Li
- Key Laboratory of Environment Pollution Monitoring and Disease Control , Ministry of Education; Department of Toxicology , School of Public Health , Guizhou Medical University , Guiyang , Guizhou 550025 , China . ; ; Tel: +86 851 8841 6172
| | - Ai-Hua Zhang
- Key Laboratory of Environment Pollution Monitoring and Disease Control , Ministry of Education; Department of Toxicology , School of Public Health , Guizhou Medical University , Guiyang , Guizhou 550025 , China . ; ; Tel: +86 851 8841 6172
| |
Collapse
|
38
|
Abstract
In the context of breast cancer, the importance of the skeleton in the regulation of primary tumour development and as a site for subsequent metastasis is well characterized. Our understanding of the contributions made by the host bone and bone marrow cells increasingly demonstrates the extent of the interaction between tumour cells and normal host cells. As a result, the need to develop and utilize therapies that can impede the growth and/or function of tumour cells while sparing normal host bone and bone marrow cells is immense and expanding. The need for these new treatments is, however, superimposed on the orthopaedic management of patients' quality of life, where pain control and continued locomotion are paramount. Indeed, the majority of the anticancer therapies used to date often result in direct or indirect damage to bone. Thus, although the bone microenvironment regulates tumour cell growth in bone, cells within the bone marrow niche also mediate many of the orthopaedic consequences of tumour progression as well as resistance to the antitumour effects of existing therapies. In this Review, we highlight the effects of existing cancer treatments on bone and the bone marrow microenvironment as well as the mechanisms mediating these effects and the current utility of modern orthopaedic interventions.
Collapse
Affiliation(s)
- Issam Makhoul
- Department of Medicine, Division of Haematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Corey O Montgomery
- Department of Orthopaedic Surgery, Centre for Orthopaedic Research, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Dana Gaddy
- Department of Physiology and Biophysics, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Larry J Suva
- Department of Orthopaedic Surgery, Centre for Orthopaedic Research, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| |
Collapse
|
39
|
The Effect of Long-Term Exercise on the Production of Osteoclastogenic and Antiosteoclastogenic Cytokines by Peripheral Blood Mononuclear Cells and on Serum Markers of Bone Metabolism. J Osteoporos 2016; 2016:5925380. [PMID: 27642534 PMCID: PMC5013218 DOI: 10.1155/2016/5925380] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/15/2016] [Accepted: 07/27/2016] [Indexed: 12/30/2022] Open
Abstract
Although it is recognized that the mechanical stresses associated with physical activity augment bone mineral density and improve bone quality, our understanding of how exercise modulates bone homeostasis at the molecular level is lacking. In a before and after trial involving 43 healthy adults, we measured the effect of six months of supervised exercise training on the spontaneous and phytohemagglutinin-induced production of osteoclastogenic cytokines (interleukin-1α, tumor necrosis factor-α), antiosteoclastogenic cytokines (transforming growth factor-β1 and interleukins 4 and 10), pleiotropic cytokines with variable effects on osteoclastogenesis (interferon-γ, interleukin-6), and T cell growth and differentiation factors (interleukins 2 and 12) by peripheral blood mononuclear cells. We also measured lymphocyte phenotypes and serum markers of bone formation (osteocalcin), bone resorption (C-terminal telopeptides of Type I collagen), and bone homeostasis (25 (OH) vitamin D, estradiol, testosterone, parathyroid hormone, and insulin-like growth factor 1). A combination of aerobic, resistance, and flexibility exercises done on average of 2.5 hours a week attenuated the production of osteoclastogenic cytokines and enhanced the production of antiosteoclastogenic cytokines. These changes were accompanied by a 16% reduction in collagen degradation products and a 9.8% increase in osteocalcin levels. We conclude that long-term moderate intensity exercise exerts a favorable effect on bone resorption by changing the balance between blood mononuclear cells producing osteoclastogenic cytokines and those producing antiosteoclastogenic cytokines. This trial is registered with Clinical Trials.gov Identifier: NCT02765945.
Collapse
|
40
|
Roosenhoff R, Anfasa F, Martina B. The pathogenesis of chronic chikungunya: evolving concepts. Future Virol 2016. [DOI: 10.2217/fvl.15.107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Chikungunya virus (CHIKV) re-emerged and caused an outbreak in the Caribbean and the Americas. CHIKV can cause incapacitating arthralgia, which may be evolved in chronic arthritis that is similar to rheumatoid arthritis that lasts for months or years. This review provides an overview of known and hypothesized mechanisms that CHIKV uses to promote chronic arthritis. We hypothesized that the chronic inflammatory response that is stimulated by persisting CHIKV replication in the joints results in the arthritic symptoms seen in patients. Most hypotheses proposed in this review need to be tested or confirmed, which may help in the development of new specific treatments and vaccines against CHIKV that will not only combat viral persistence but also prevent tissue damage.
Collapse
Affiliation(s)
- Rueshandra Roosenhoff
- ARTEMIS One Health Research Institute, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
- Curacao Biomedical & Health Research Institute, Curacao
| | - Fatih Anfasa
- Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Byron Martina
- ARTEMIS One Health Research Institute, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
41
|
Faienza MF, Ventura A, Colucci S, Cavallo L, Grano M, Brunetti G. Bone Fragility in Turner Syndrome: Mechanisms and Prevention Strategies. Front Endocrinol (Lausanne) 2016; 7:34. [PMID: 27199891 PMCID: PMC4844601 DOI: 10.3389/fendo.2016.00034] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 04/11/2016] [Indexed: 01/18/2023] Open
Abstract
Bone fragility is recognized as one of the major comorbidities in Turner syndrome (TS). The mechanisms underlying bone impairment in affected patients are not clearly elucidated, but estrogen deficiency and X-chromosomal abnormalities represent important factors. Moreover, although many girls with TS undergo recombinant growth hormone therapy to treat short stature, the efficacy of this treatment on bone mineral density is controversial. The present review will focus on bone fragility in subjects with TS, providing an overview on the pathogenic mechanisms and some prevention strategies.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Pediatrics Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “A. Moro”, Bari, Italy
| | - Annamaria Ventura
- Pediatrics Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “A. Moro”, Bari, Italy
| | - Silvia Colucci
- Section of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari “A. Moro”, Bari, Italy
| | - Luciano Cavallo
- Pediatrics Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “A. Moro”, Bari, Italy
| | - Maria Grano
- Department of Emergency and Organ Transplantation (DETO), University of Bari “A. Moro”, Bari, Italy
| | - Giacomina Brunetti
- Section of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari “A. Moro”, Bari, Italy
- *Correspondence: Giacomina Brunetti,
| |
Collapse
|