1
|
Wu Y, Song P, Wang M, Liu H, Jing Y, Su J. Extracellular derivatives for bone metabolism. J Adv Res 2024; 66:329-347. [PMID: 38218580 DOI: 10.1016/j.jare.2024.01.011] [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: 11/03/2023] [Revised: 12/13/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Bone metabolism can maintain the normal homeostasis and function of bone tissue. Once the bone metabolism balance is broken, it will cause osteoporosis, osteoarthritis, bone defects, bone tumors, or other bone diseases. However, such orthopedic diseases still have many limitations in clinical treatment, such as drug restrictions, drug tolerance, drug side effects, and implant rejection. AIM OF REVIEW In complex bone therapy and bone regeneration, extracellular derivatives have become a promising research focus to solve the problems of bone metabolic diseases. These derivatives, which include components such as extracellular matrix, growth factors, and extracellular vesicles, have significant therapeutic potential. It has the advantages of good biocompatibility, low immune response, and dynamic demand for bone tissue. The purpose of this review is to provide a comprehensive perspective on extracellular derivatives for bone metabolism and elucidate the intrinsic properties and versatility of extracellular derivatives. Further discussion of them as innovative advanced orthopedic materials for improving the effectiveness of bone therapy and regeneration processes. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, we first listed the types and functions of three extracellular derivatives. Then, we discussed the effects of extracellular derivatives of different cell sources on bone metabolism. Subsequently, we collected applications of extracellular derivatives in the treatment of bone metabolic diseases and summarized the advantages and challenges of extracellular derivatives in clinical applications. Finally, we prospected the extracellular derivatives in novel orthopedic materials and clinical applications. We hope that the comprehensive understanding of extracellular derivatives in bone metabolism will provide new solutions to bone diseases.
Collapse
Affiliation(s)
- Yan Wu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Peiran Song
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Miaomiao Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Department of Rehabilitation Medicine, Shanghai Zhongye Hospital, Shanghai 200941, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China.
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
2
|
Ma H, Cai X, Hu J, Song S, Zhu Q, Zhang Y, Ma R, Shen D, Yang W, Zhou P, Zhang D, Luo Q, Hong J, Li N. Association of systemic inflammatory response index with bone mineral density, osteoporosis, and future fracture risk in elderly hypertensive patients. Postgrad Med 2024; 136:406-416. [PMID: 38753519 DOI: 10.1080/00325481.2024.2354158] [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: 04/01/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVES This study sought to investigate the relationship between the systemic inflammatory response index (SIRI) and bone mineral density (BMD), osteoporosis, and future fracture risk in elderly hypertensive patients. METHODS Elderly hypertensive patients (age ≥60 years) who attended our hospital between January 2021 and December 2023 and completed BMD screening were included in the study. Analyses were performed with multivariate logistic and linear regression. RESULTS The multiple linear regression indicated that SIRI levels were significantly negatively correlated with lumbar 1 BMD (β = -0.15, 95% CI: -0.24, -0.05), lumbar 2 BMD (β = -0.15, 95% CI: -0.24, -0.05), lumbar 3 BMD (β = -1.35, 95% CI: -0.23, -0.02), lumbar 4 BMD (β = -0.11, 95% CI: -0.30, -0.10), femur neck BMD (β = -0.11, 95% CI: -0.18, -0.05) and Ward's triangle BMD (β = -0.12, 95% CI: -0.20, -0.05) among elderly hypertensive patients, after fully adjusting for confounders. Furthermore, we observed that SIRI was positively associated with future fracture risk in elderly hypertensive patients. Specifically, SIRI was associated with an increased risk of major osteoporotic fractures (β = 0.33) and hip fractures (β = 0.25). The logistic regression analysis indicated that there is an association between the SIRI level and an increased risk of osteoporosis (OR = 1.60, 95% CI = 1.37, 1.87), after fully adjusting for confounders. CONCLUSIONS Our findings indicate a potential association between SIRI and BMD, osteoporosis, and the risk of future fractures in elderly hypertensive patients. However, further studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Huimin Ma
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Xintian Cai
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Junli Hu
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Shuaiwei Song
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Qing Zhu
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Yingying Zhang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Rui Ma
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Di Shen
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Wenbo Yang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Pan Zhou
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Delian Zhang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Qin Luo
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Jing Hong
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Nanfang Li
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| |
Collapse
|
3
|
Mo S, Jang JS, Lee SH, Kim HH. Single-cell transcriptome analysis reveals periodontal ligament fibroblast heterogeneity with distinct IL-1β and RANKL expression in periodontitis. Mol Cells 2024; 47:100059. [PMID: 38554844 PMCID: PMC11026731 DOI: 10.1016/j.mocell.2024.100059] [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: 02/19/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024] Open
Abstract
Periodontitis (PD) is an inflammatory disease with alveolar bone destruction by osteoclasts (OCs). In PD, both inflammation and OC activation are significantly influenced by periodontal ligament fibroblasts (PDL-Fib). Yet, whether PDL-Fib has heterogeneity and whether distinct PDL-Fib subsets have specific functions have not been investigated. In this study, we discovered the complexity of PDL-Fib in PD, utilizing single-cell RNA sequencing data from human PD patients. We identified distinct subpopulations of PDL-Fib: one expressing interleukin-1 beta (IL-1β) and another expressing the receptor activator of nuclear factor-kappa B ligand (RANKL), both crucial in OC differentiation and bone resorption. In periodontal tissues of mice with PD, active IL-1β, cleaved caspase 1, and nucleotide-binding oligomerization domain-like receptor 3 (NLPR3) were significantly elevated, implicating the NLRP3 inflammasome in IL-1β production. Upon stimulation of PDL-Fib with LPS from Porphyromonas gingivalis (pg), the most well-characterized periodontal bacteria, a more rapid increase in IL-1β, followed by RANKL induction, was observed. IL-1β and tumor necrosis factor alpha (TNF-α), another LPS-responsive cytokine, effectively increased RANKL in PDL-Fib, suggesting an indirect effect of pgLPS through IL-1β and TNF-α on RANKL induction. Immunohistological analyses of mouse periodontal tissues also showed markedly elevated levels of IL-1β and RANKL upon PD induction and displayed separate locations of IL-1β-expressing PDL-Fib and RANKL-expressing PDL-Fib in PD. The heterogenic feature of fibroblasts expressing IL-1β and RANKL was also mirrored in our combined cross-tissue single-cell RNA sequencing datasets analysis. In summary, our study elucidates the heterogeneity of PDL-Fib, highlighting distinct functional groups for producing RANKL and IL-1β, which collectively promote OC generation and bone destruction in PD.
Collapse
Affiliation(s)
- Shenzheng Mo
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Ji Sun Jang
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Seung Hye Lee
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Hong-Hee Kim
- Department of Cell and Developmental Biology, Dental Research Institute, School of Dentistry, Seoul National University, Seoul 03080, Korea.
| |
Collapse
|
4
|
Engelmann J, Ragipoglu D, Ben-Batalla I, Loges S. The Role of TAM Receptors in Bone. Int J Mol Sci 2023; 25:233. [PMID: 38203403 PMCID: PMC10779100 DOI: 10.3390/ijms25010233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
The TAM (TYRO3, MERTK, and AXL) family of receptor tyrosine kinases are pleiotropic regulators of adult tissue homeostasis maintaining organ integrity and self-renewal. Disruption of their homeostatic balance fosters pathological conditions like autoinflammatory or degenerative diseases including rheumatoid arthritis, lupus erythematodes, or liver fibrosis. Moreover, TAM receptors exhibit prominent cell-transforming properties, promoting tumor progression, metastasis, and therapy resistance in various cancer entities. Emerging evidence shows that TAM receptors are involved in bone homeostasis by regulating osteoblastic bone formation and osteoclastic bone resorption. Therefore, TAM receptors emerge as new key players of the regulatory cytokine network of osteoblasts and osteoclasts and represent accessible targets for pharmacologic therapy for a broad set of different bone diseases, including primary and metastatic bone tumors, rheumatoid arthritis, or osteoporosis.
Collapse
Affiliation(s)
- Janik Engelmann
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, 68167 Mannheim, Germany; (D.R.); (I.B.-B.)
- Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Deniz Ragipoglu
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, 68167 Mannheim, Germany; (D.R.); (I.B.-B.)
- Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Isabel Ben-Batalla
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, 68167 Mannheim, Germany; (D.R.); (I.B.-B.)
- Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Sonja Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, 68167 Mannheim, Germany; (D.R.); (I.B.-B.)
- Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
5
|
Chen C, Wang S, Wang N, Zheng Y, Zhou J, Hong M, Chen Z, Wang S, Wang Z, Xiang S. Icariin inhibits prostate cancer bone metastasis and destruction via suppressing TAM/CCL5-mediated osteoclastogenesis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155076. [PMID: 37716031 DOI: 10.1016/j.phymed.2023.155076] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 09/04/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND Bone metastasis occurs in nearly 70% of patients with metastatic prostate cancer (PCa), and represents the leading cause of death in patients with PCa. Emerging evidence has demonstrated the potential activities of icariin in modulating bone metabolism and remodelling the tumor microenvironment (TME). However, whether icariin could inhibit PCa bone metastasis and destruction by modulating the TME as well as the underlying mechanisms remains unclear. PURPOSE This study investigated whether icariin could inhibit PCa bone metastasis and destruction by modulating the bone TME as well as the underlying mechanisms. METHODS Osteoclasts were induced from mouse bone marrow-derived macrophages (BMMs) or Raw264.7 cells. PCa cells were cultured in the conditional medium (CM) of macrophages in vitro or co-injected with macrophages in vivo to simulate their coexistence in the TME. Multiple molecular biology experiments and the mouse RM1-Luc PCa bone metastasis model were used to explore the inhibitory activity and mechanism of icariin on PCa metastasis and bone destruction. RESULTS Icariin treatment significantly suppressed PCa growth, bone metastasis and destruction as well as osteoclastogenesis in vivo. Furthermore, icariin remarkably inhibited osteoclast differentiation, even in the presence of the CM of tumor-associated macrophages (TAMs), while exhibiting no obvious effect on osteoblasts. Moreover, icariin suppressed the M2 phenotype polarization of Raw264.7-derived TAMs and transcriptionally attenuated their CC motif chemokine ligand 5 (CCL5) expression and secretion via inhibiting SPI1. Additionally, CCL5 induced the differentiation and chemotaxis of osteoclast precursor cells by binding with its receptor CCR5. The clinicopathological analysis further verified the positive correlation between the TAM/CCL5/CCR5 axis and osteoclastogenesis within the TME of PCa patients. More importantly, icariin remarkably suppressed PCa metastasis-induced bone destruction in vivo by inhibiting osteoclastogenesis via downregulating the TAM/CCL5 pathway. CONCLUSION Altogether, these results not only implicate icariin as a promising candidate immunomodulator for PCa bone metastasis and destruction but also shed novel insight into targeting TAM/CCL5-mediated osteoclastogenesis as a potential treatment strategy for osteolytic bone metastasis. This study helps to advance the understanding of the crosstalk between bone TME and bone homeostasis.
Collapse
Affiliation(s)
- Chiwei Chen
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shengqi Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Neng Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yifeng Zheng
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianfu Zhou
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Min Hong
- Department of Pathology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiqiang Chen
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shusheng Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiyu Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Songtao Xiang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
6
|
Choa R, Panaroni C, Bhatia R, Raje N. It is worth the weight: obesity and the transition from monoclonal gammopathy of undetermined significance to multiple myeloma. Blood Adv 2023; 7:5510-5523. [PMID: 37493975 PMCID: PMC10515310 DOI: 10.1182/bloodadvances.2023010822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/05/2023] [Accepted: 07/05/2023] [Indexed: 07/27/2023] Open
Abstract
The overweight/obesity epidemic is a serious public health concern that affects >40% of adults globally and increases the risk of numerous chronic diseases, such as type 2 diabetes, heart disease, and various cancers. Multiple myeloma (MM) is a lymphohematopoietic cancer caused by the uncontrolled clonal expansion of plasma cells. Recent studies have shown that obesity is a risk factor not only for MM but also monoclonal gammopathy of undetermined significance (MGUS), a precursor disease state of MM. Furthermore, obesity may promote the transition from MGUS to MM. Thus, in this review, we summarize the epidemiological evidence regarding the role of obesity in MM and MGUS, discuss the biologic mechanisms that drive these disease processes, and detail the obesity-targeted pharmacologic and lifestyle interventions that may reduce the risk of progression from MGUS to MM.
Collapse
Affiliation(s)
- Ruth Choa
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Cristina Panaroni
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Roma Bhatia
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Noopur Raje
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
7
|
Zhang Y, Liu D, Vithran DTA, Kwabena BR, Xiao W, Li Y. CC chemokines and receptors in osteoarthritis: new insights and potential targets. Arthritis Res Ther 2023; 25:113. [PMID: 37400871 PMCID: PMC10316577 DOI: 10.1186/s13075-023-03096-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/23/2023] [Indexed: 07/05/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease accompanied by the activation of innate and adaptive immune systems-associated inflammatory responses. Due to the local inflammation, the expression of various cytokines was altered in affected joints, including CC motif chemokine ligands (CCLs) and their receptors (CCRs). As essential members of chemokines, CCLs and CCRs played an important role in the pathogenesis and treatment of OA. The bindings between CCLs and CCRs on the chondrocyte membrane promoted chondrocyte apoptosis and the release of multiple matrix-degrading enzymes, which resulted in cartilage degradation. In addition, CCLs and CCRs had chemoattractant functions to attract various immune cells to osteoarthritic joints, further leading to the aggravation of local inflammation. Furthermore, in the nerve endings of joints, CCLs and CCRs, along with several cellular factors, contributed to pain hypersensitivity by releasing neurotransmitters in the spinal cord. Given this family's diverse and complex functions, targeting the functional network of CCLs and CCRs is a promising strategy for the prognosis and treatment of OA in the future.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | | | - Bosomtwe Richmond Kwabena
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
8
|
Han L, Zhang L. CCL21/CCR7 axis as a therapeutic target for autoimmune diseases. Int Immunopharmacol 2023; 121:110431. [PMID: 37331295 DOI: 10.1016/j.intimp.2023.110431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/20/2023]
Abstract
Chemokine receptor 7 (CCR7) is a G protein-coupled receptor containing 7 transmembrane domains that is expressed on various cells, such as naive T/B cells, central memory T cells, regulatory T cells, immature/mature dendritic cells (DCs), natural killer cells, and a minority of tumor cells. Chemokine ligand 21 (CCL21) is the known high-affinity ligand that binds to CCR7 and drives cell migration in tissues. CCL21 is mainly produced by stromal cells and lymphatic endothelial cells, and its expression is significantly increased under inflammatory conditions. Genome-wide association studies (GWAS) have shown a strong association between CCL21/CCR7 axis and disease severity in patients with rheumatoid arthritis, sjogren's syndrome, systemic lupus erythematosus, polymyositis, ankylosing spondylitis, and asthma. Disrupting CCL21/CCR7 interaction with antibodies or inhibitors prevents the migration of CCR7-expressing immune and non-immune cells at the site of inflammation and reduces disease severity. This review emphasizes the importance of the CCL21 /CCR7 axis in autoimmune diseases and evaluates its potential as a novel therapeutic target for these conditions.
Collapse
Affiliation(s)
- Le Han
- Department of Pharmacy, The Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin 214400, China
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
9
|
Cellular and Molecular Mechanisms Associating Obesity to Bone Loss. Cells 2023; 12:cells12040521. [PMID: 36831188 PMCID: PMC9954309 DOI: 10.3390/cells12040521] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Obesity is an alarming disease that favors the upset of other illnesses and enhances mortality. It is spreading fast worldwide may affect more than 1 billion people by 2030. The imbalance between excessive food ingestion and less energy expenditure leads to pathological adipose tissue expansion, characterized by increased production of proinflammatory mediators with harmful interferences in the whole organism. Bone tissue is one of those target tissues in obesity. Bone is a mineralized connective tissue that is constantly renewed to maintain its mechanical properties. Osteoblasts are responsible for extracellular matrix synthesis, while osteoclasts resorb damaged bone, and the osteocytes have a regulatory role in this process, releasing growth factors and other proteins. A balanced activity among these actors is necessary for healthy bone remodeling. In obesity, several mechanisms may trigger incorrect remodeling, increasing bone resorption to the detriment of bone formation rates. Thus, excessive weight gain may represent higher bone fragility and fracture risk. This review highlights recent insights on the central mechanisms related to obesity-associated abnormal bone. Publications from the last ten years have shown that the main molecular mechanisms associated with obesity and bone loss involve: proinflammatory adipokines and osteokines production, oxidative stress, non-coding RNA interference, insulin resistance, and changes in gut microbiota. The data collection unveils new targets for prevention and putative therapeutic tools against unbalancing bone metabolism during obesity.
Collapse
|
10
|
Kim Y, Kim GT. Positive Effects of Biologics on Osteoporosis in Rheumatoid Arthritis. JOURNAL OF RHEUMATIC DISEASES 2023; 30:3-17. [PMID: 37476528 PMCID: PMC10351356 DOI: 10.4078/jrd.22.0046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/10/2022] [Accepted: 12/10/2022] [Indexed: 07/22/2023]
Abstract
Osteoporosis is a systemic skeletal disorder that causes vulnerability of bones to fracture owing to reduction in bone density and deterioration of the bone tissue microstructure. The prevalence of osteoporosis is higher in patients with autoimmune inflammatory rheumatic diseases, including rheumatoid arthritis (RA), than in those of the general population. In this autoimmune inflammatory rheumatic disease, in addition to known risk factors for osteoporosis, various factors such as chronic inflammation, autoantibodies, metabolic disorders, drugs, and decreased physical activity contribute to additional risk. In RA, disease-related inflammation plays an important role in local or systemic bone loss, and active treatment for inflammation can help prevent osteoporosis. In addition to conventional synthetic disease-modifying anti-rheumatic drugs that have been traditionally used for treatment of RA, biologic DMARDs and targeted synthetic DMARDs have been widely used. These agents can be employed more selectively and precisely based on disease pathogenesis. It has been reported that these drugs can inhibit bone loss by not only reducing inflammation in RA, but also by inhibiting bone resorption and promoting bone formation. In this review, the pathogenesis and research results of the increase in osteoporosis in RA are reviewed, and the effects of biological agents on osteoporosis are discussed.
Collapse
Affiliation(s)
- Yunkyung Kim
- Division of Rheumatology, Department of Internal Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Korea
| | - Geun-Tae Kim
- Division of Rheumatology, Department of Internal Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Korea
| |
Collapse
|
11
|
Murayama MA, Shimizu J, Miyabe C, Yudo K, Miyabe Y. Chemokines and chemokine receptors as promising targets in rheumatoid arthritis. Front Immunol 2023; 14:1100869. [PMID: 36860872 PMCID: PMC9968812 DOI: 10.3389/fimmu.2023.1100869] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that commonly causes inflammation and bone destruction in multiple joints. Inflammatory cytokines, such as IL-6 and TNF-α, play important roles in RA development and pathogenesis. Biological therapies targeting these cytokines have revolutionized RA therapy. However, approximately 50% of the patients are non-responders to these therapies. Therefore, there is an ongoing need to identify new therapeutic targets and therapies for patients with RA. In this review, we focus on the pathogenic roles of chemokines and their G-protein-coupled receptors (GPCRs) in RA. Inflamed tissues in RA, such as the synovium, highly express various chemokines to promote leukocyte migration, tightly controlled by chemokine ligand-receptor interactions. Because the inhibition of these signaling pathways results in inflammatory response regulation, chemokines and their receptors could be promising targets for RA therapy. The blockade of various chemokines and/or their receptors has yielded prospective results in preclinical trials using animal models of inflammatory arthritis. However, some of these strategies have failed in clinical trials. Nonetheless, some blockades showed promising results in early-phase clinical trials, suggesting that chemokine ligand-receptor interactions remain a promising therapeutic target for RA and other autoimmune diseases.
Collapse
Affiliation(s)
- Masanori A Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| | - Jun Shimizu
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Chie Miyabe
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazuo Yudo
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Yoshishige Miyabe
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| |
Collapse
|
12
|
Phan QT, Chua KY, Jin A, Winkler C, Koh WP. CXCL9 Predicts the Risk of Osteoporotic Hip Fracture in a Prospective Cohort of Chinese Men-A Matched Case-Control Study. J Bone Miner Res 2022; 37:1843-1849. [PMID: 35810382 PMCID: PMC9804917 DOI: 10.1002/jbmr.4646] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/03/2022] [Accepted: 07/05/2022] [Indexed: 01/07/2023]
Abstract
Recent experimental work has identified CXCL9 as a promoter for the differentiation of osteoclast progenitors into osteoclasts, with resultant bone resorption. However, no human study has validated an association between this chemokine and osteoporosis or fracture risk. We conducted a matched case-control study nested in the prospective, population-based Singapore Chinese Health Study. Fifty-five men and 119 women with incident hip fractures, occurring median 6.2 years after blood collection, were matched individually to controls by age at recruitment, sex, and duration of blood storage. Serum chemokines, CXCL9 and CXCL10, were measured using immunoassays. Multivariable conditional logistic regression models that included age at blood collection, body mass index, smoking, and diabetes as covariates were used to estimate odds ratios (OR) and 95% confidence intervals (CI) for association with hip fracture risk. Predictive utility of chemokine for hip fracture risk was examined by comparing area under receiver operating characteristic curves (AUC) between prognostic models with and without the chemokine. Increasing CXCL9 levels were associated with increasing hip fracture risk in men but not in women (pinteraction = 0.002); comparing extreme quartiles, the OR (95% CI) in the highest quartile was 10.35 (1.90-56.39) in men (ptrend = 0.002) but 1.46 (0.59-3.60) in women (ptrend = 0.32). Adding CXCL9 to a prognostic model that already incorporated age and other risk factors improved the AUC (95% CI) from 0.65 (0.55-0.76) to 0.74 (0.65-0.83) for the predictive utility of hip fractures in men but not in women. Conversely, the association between CXCL10 and hip fracture risk was not statistically significant in either sex. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Quang Tien Phan
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore.,Centre for Bioimaging Sciences, National University of Singapore, Singapore, Singapore
| | - Kevin Yiqiang Chua
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - Aizhen Jin
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christoph Winkler
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore.,Centre for Bioimaging Sciences, National University of Singapore, Singapore, Singapore
| | - Woon-Puay Koh
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
13
|
Chen B, Liang Y, Song Y, Liang Y, Jiao J, Bai H, Li Y. Photothermal-Controlled Release of IL-4 in IL-4/PDA-Immobilized Black Titanium Dioxide (TiO 2) Nanotubes Surface to Enhance Osseointegration: An In Vivo Study. MATERIALS (BASEL, SWITZERLAND) 2022; 15:5962. [PMID: 36079344 PMCID: PMC9457063 DOI: 10.3390/ma15175962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
Host immune response has gradually been accepted as a critical factor in achieving successful implant osseointegration. The aim of this study is to create a favorable immune microenvironment by the dominant release of IL-4 during the initial few days after implant insertion to mitigate early inflammatory reactions and facilitate osseointegration. Herein, the B-TNT/PDA/IL-4 substrate was established by immobilizing an interleukin-4 (IL-4)/polydopamine (PDA) coating on a black TiO2 nanotube (B-TNT) surface, achieving on-demand IL-4 release under near infrared (NIR) irradiation. Gene Ontology (GO) enrichment analyses based on high-throughput DNA microarray data revealed that IL-4 addition inhibited osteoclast differentiation and function. Animal experiment results suggested that the B-TNT/PDA/IL-4+Laser substrate induced the least inflammatory, tartrate-resistant acid phosphatase, inducible nitric oxide synthase and the most CD163 positive cells, compared to the Ti group at 7 days post-implantation. In addition, 28 days post-implantation, micro-computed tomography results showed the highest bone volume/total volume, trabecular thickness, trabecular number and the lowest trabecular separation, while Hematoxylin-eosin and Masson-trichrome staining revealed the largest amount of new bone formation for the B-TNT/PDA/IL-4+Laser group. This study revealed the osteoimmunoregulatory function of the novel B-TNT/PDA/IL-4 surface by photothermal release of IL-4 at an early period post-implantation, thus paving a new way for dental implant surface modification.
Collapse
Affiliation(s)
- Bo Chen
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| | - Yu Liang
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| | - Yunjia Song
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| | - Yunkai Liang
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| | - Jian Jiao
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| | - Hong Bai
- Tianjin Key Laboratory of Cellular and Molecular Immunology and Key Laboratory of the Educational Ministry of China, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Ying Li
- School of Dentistry, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
14
|
Ding D, Wang L, Yan J, Zhou Y, Feng G, Ma L, Yang Y, Pei X, Jin Q. Zoledronic acid generates a spatiotemporal effect to attenuate osteoarthritis by inhibiting potential Wnt5a-associated abnormal subchondral bone resorption. PLoS One 2022; 17:e0271485. [PMID: 35900969 PMCID: PMC9333271 DOI: 10.1371/journal.pone.0271485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022] Open
Abstract
This study aimed to determine the effects of zoledronic acid (ZOL) on OA in rats and explored the molecular mechanism of osteoclast activation in early OA. A knee OA rat model was designed by surgically destabilizing the medial meniscus (DMM). Seventy-two male rats were randomly assigned to Sham+phosphate-buffered saline (PBS), DMM+PBS, and DMM+ZOL groups; rats were administered with 100 μg/Kg ZOL or PBS, twice weekly for 4 weeks. After 2, 4, 8, and 12 weeks of OA induction, the thickness of the hyaline and calcified cartilage layers was calculated using hematoxylin and eosin staining, degenerated cartilage stained with Safranin O-fast green staining was evaluated and scored, tartrate-resistant acid phosphatase (TRAP)-stained osteoclasts were counted, changes in subchondral bone using micro-computed tomography were analyzed, and PINP and CTX-I levels were detected using enzyme-linked immunosorbent assay. Using these results, 18 male rats were randomly assigned to three groups. Four weeks after surgery, Wnt5a, RANKL, CXCL12, and NFATc1 protein levels were measured in subchondral bone using western blotting, and mRNA levels of genes related to osteoclastogenesis in subchondral bone were measured using quantitative polymerase chain reaction. Bone marrow-derived macrophages were isolated as osteoclast precursors, and cell differentiation, migration, and adhesion were assessed by TRAP staining and Transwell assays, revealing that DMM induced knee OA in rats. Progressive cartilage loss was observed 12 weeks after OA induction. Subchondral bone remodeling was dominated by bone resorption during early OA (within 4 weeks), whereas bone formation was increased 8 weeks later. ZOL suppressed bone resorption by inhibiting Wnt5a signaling in early OA, improved the imbalance of subchondral bone remodeling, reduced cartilage degeneration, and delayed OA progression. Additionally, ZOL delayed OA progression and reduced cartilage degeneration via a spatiotemporal effect in DMM-induced OA. Osteoclast activity in early OA might be associated with Wnt5a signaling, indicating a possible novel strategy for OA treatment.
Collapse
Affiliation(s)
- Dong Ding
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Clinical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
- Hand & Ankle Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, P.R. China
- * E-mail: (QJ); (DD)
| | - Limei Wang
- Medical College, Qingdao Binhai University, Qingdao, Shandong, P.R. China
| | - Jiangbo Yan
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Clinical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Yong Zhou
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Clinical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Gangning Feng
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Clinical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Long Ma
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Yong Yang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Clinical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Xiuying Pei
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Clinical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Qunhua Jin
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
- * E-mail: (QJ); (DD)
| |
Collapse
|
15
|
C‑X‑C receptor 7 agonist acts as a C‑X‑C motif chemokine ligand 12 inhibitor to ameliorate osteoclastogenesis and bone resorption. Mol Med Rep 2022; 25:78. [PMID: 35014674 PMCID: PMC8778739 DOI: 10.3892/mmr.2022.12594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/09/2021] [Indexed: 12/03/2022] Open
Abstract
The C-X-C receptor (CXCR) 7 agonist, VUF11207, is a chemical compound that binds specifically to CXCR7, and negatively regulates C-X-C motif chemokine ligand 12 (CXCL12) and CXCR4-induced cellular events. Lipopolysaccharide (LPS) can induce inflammatory cytokines and pathological bone loss. LPS also induces expression of CXCL12, enhancing sensitivity to receptor activator of NF-κB ligand (RANKL) and tumor necrosis factor-α (TNF-α) in vivo. RANKL and TNF-α induce the differentiation of osteoclasts into osteoclast precursors and bone resorption. The current study was performed to examine the effects of a CXCR7 agonist on osteoclastogenesis and bone resorption induced by LPS in vivo. In addition, the mechanisms underlying these in vivo effects were investigated by in vitro experiments. Eight-week-old male C57BL/6J mice were subcutaneously injected over the calvariae with LPS alone or LPS and CXCR7 agonist. After sacrifice, the number of osteoclasts and the bone resorption area were measured. In vitro experiments were performed to investigate the effects of CXCL12 and CXCR7 agonist on osteoclastogenesis induced by RANKL and TNF-α. Mice injected with LPS and CXCR7 agonist showed significantly reduced osteoclastogenesis and bone resorption compared with mice injected with LPS alone. Moreover, the CXCR7 agonist inhibited CXCL12 enhancement of RANKL- and TNF-α-induced osteoclastogenesis in vitro. Thus, CXCR7 agonist inhibited LPS-induced osteoclast-associated cytokines, such as RANKL and TNF-α, as well as RANKL- and TNF-α-induced osteoclastogenesis in vitro by modulating CXCL12-mediated enhancement of osteoclastogenesis. In conclusion, CXCR7 agonist reduced CXCL12-mediated osteoclastogenesis and bone resorption.
Collapse
|
16
|
Zhou X, Yuan W, Xiong X, Zhang Z, Liu J, Zheng Y, Wang J, Liu J. HO-1 in Bone Biology: Potential Therapeutic Strategies for Osteoporosis. Front Cell Dev Biol 2021; 9:791585. [PMID: 34917622 PMCID: PMC8669958 DOI: 10.3389/fcell.2021.791585] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/12/2021] [Indexed: 02/05/2023] Open
Abstract
Osteoporosis is a prevalent bone disorder characterized by bone mass reduction and deterioration of bone microarchitecture leading to bone fragility and fracture risk. In recent decades, knowledge regarding the etiological mechanisms emphasizes that inflammation, oxidative stress and senescence of bone cells contribute to the development of osteoporosis. Studies have demonstrated that heme oxygenase 1 (HO-1), an inducible enzyme catalyzing heme degradation, exhibits anti-inflammatory, anti-oxidative stress and anti-apoptosis properties. Emerging evidence has revealed that HO-1 is critical in the maintenance of bone homeostasis, making HO-1 a potential target for osteoporosis treatment. In this Review, we aim to provide an introduction to current knowledge of HO-1 biology and its regulation, focusing specifically on its roles in bone homeostasis and osteoporosis. We also examine the potential of HO-1-based pharmacological therapeutics for osteoporosis and issues faced during clinical translation.
Collapse
Affiliation(s)
- Xueman Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Wenxiu Yuan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xiong
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhenzhen Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yingcheng Zheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jin Liu
- Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Ramser A, Greene E, Wideman R, Dridi S. Local and Systemic Cytokine, Chemokine, and FGF Profile in Bacterial Chondronecrosis with Osteomyelitis (BCO)-Affected Broilers. Cells 2021; 10:3174. [PMID: 34831397 PMCID: PMC8620240 DOI: 10.3390/cells10113174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022] Open
Abstract
Complex disease states, like bacterial chondronecrosis with osteomyelitis (BCO), not only result in physiological symptoms, such as lameness, but also a complex systemic reaction involving immune and growth factor responses. For the modern broiler (meat-type) chickens, BCO is an animal welfare, production, and economic concern involving bacterial infection, inflammation, and bone attrition with a poorly defined etiology. It is, therefore, critical to define the key inflammatory and bone-related factors involved in BCO. In this study, the local bone and systemic blood profile of inflammatory modulators, cytokines, and chemokines was elucidated along with inflammasome and key FGF genes. BCO-affected bone showed increased expression of cytokines IL-1β, while BCO-affected blood expressed upregulated TNFα and IL-12. The chemokine profile revealed increased IL-8 expression in both BCO-affected bone and blood in addition to inflammasome NLRC5 being upregulated in circulation. The key FGF receptor, FGFR1, was significantly downregulated in BCO-affected bone. The exposure of two different bone cell types, hFOB and chicken primary chondrocytes, to plasma from BCO-affected birds, as well as recombinant TNFα, resulted in significantly decreased cell viability. These results demonstrate an expression of proinflammatory and bone-resorptive factors and their potential contribution to BCO etiology through their impact on bone cell viability. This unique profile could be used for improved non-invasive detection of BCO and provides potential targets for treatments.
Collapse
Affiliation(s)
- Alison Ramser
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA; (A.R.); (E.G.); (R.W.)
- Department of Poultry Science, Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA
| | - Elizabeth Greene
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA; (A.R.); (E.G.); (R.W.)
| | - Robert Wideman
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA; (A.R.); (E.G.); (R.W.)
| | - Sami Dridi
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA; (A.R.); (E.G.); (R.W.)
- Department of Poultry Science, Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
18
|
Rodríguez V, Rivoira M, Picotto G, de Barboza GD, Collin A, de Talamoni NT. Analysis of the molecular mechanisms by flavonoids with potential use for osteoporosis prevention or therapy. Curr Med Chem 2021; 29:2913-2936. [PMID: 34547992 DOI: 10.2174/0929867328666210921143644] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/11/2021] [Accepted: 08/15/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Osteoporosis is the most common skeletal disorder worldwide. Flavonoids have the potential to alleviate bone alterations in osteoporotic patients with the advantage of being safer and less expensive than the conventional therapies. OBJECTIVE The main objective is to analyze the molecular mechanisms triggered in bone by different subclasses of flavonoids. In addition, this review provides an up-to-date overview on the cellular and molecular aspects of osteoporotic bones versus healthy bones, and a brief description of some epidemiological studies indicating that flavonoids could be useful for osteoporosis treatment. METHODS The PubMed database was searched in the range of years 2001- 2021 using the keywords osteoporosis, flavonoids, and their subclasses such as flavones, flavonols, flavanols, isoflavones, flavanones and anthocyanins, focusing the data on the molecular mechanisms triggered in bone. RESULTS Although flavonoids comprise many compounds that differ in structure, their effects on bone loss in postmenopausal women or in ovariectomized-induced osteoporotic animals are quite similar. Most of them increase bone mineral density and bone strength, which occur through enhancement of osteoblastogenesis and osteoclast apoptosis, decrease in osteoclastogenesis as well as increase in neovascularization on the site of the osteoporotic fracture. CONCLUSION Several molecules of signaling pathways are involved in the effect of flavonoids on osteoporotic bone. Whether all flavonoids have a common mechanism or they act as ligands of estrogen receptors remain to be established. More clinical trials are necessary to know better their safety, efficacy, delivery and bioavailability in humans, as well as comparative studies with conventional therapies.
Collapse
Affiliation(s)
- Valeria Rodríguez
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba. Argentina
| | - María Rivoira
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba. Argentina
| | - Gabriela Picotto
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba. Argentina
| | - Gabriela Díaz de Barboza
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba. Argentina
| | - Alejandro Collin
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba. Argentina
| | - Nori Tolosa de Talamoni
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do. Piso, Ciudad Universitaria, 5000 Córdoba. Argentina
| |
Collapse
|
19
|
Saxena Y, Routh S, Mukhopadhaya A. Immunoporosis: Role of Innate Immune Cells in Osteoporosis. Front Immunol 2021; 12:687037. [PMID: 34421899 PMCID: PMC8374941 DOI: 10.3389/fimmu.2021.687037] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis or porous bone disorder is the result of an imbalance in an otherwise highly balanced physiological process known as 'bone remodeling'. The immune system is intricately involved in bone physiology as well as pathologies. Inflammatory diseases are often correlated with osteoporosis. Inflammatory mediators such as reactive oxygen species (ROS), and pro-inflammatory cytokines and chemokines directly or indirectly act on the bone cells and play a role in the pathogenesis of osteoporosis. Recently, Srivastava et al. (Srivastava RK, Dar HY, Mishra PK. Immunoporosis: Immunology of Osteoporosis-Role of T Cells. Frontiers in immunology. 2018;9:657) have coined the term "immunoporosis" to emphasize the role of immune cells in the pathology of osteoporosis. Accumulated pieces of evidence suggest both innate and adaptive immune cells contribute to osteoporosis. However, innate cells are the major effectors of inflammation. They sense various triggers to inflammation such as pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), cellular stress, etc., thus producing pro-inflammatory mediators that play a critical role in the pathogenesis of osteoporosis. In this review, we have discussed the role of the innate immune cells in great detail and divided these cells into different sections in a systemic manner. In the beginning, we talked about cells of the myeloid lineage, including macrophages, monocytes, and dendritic cells. This group of cells explicitly influences the skeletal system by the action of production of pro-inflammatory cytokines and can transdifferentiate into osteoclast. Other cells of the myeloid lineage, such as neutrophils, eosinophils, and mast cells, largely impact osteoporosis via the production of pro-inflammatory cytokines. Further, we talked about the cells of the lymphoid lineage, including natural killer cells and innate lymphoid cells, which share innate-like properties and play a role in osteoporosis. In addition to various innate immune cells, we also discussed the impact of classical pro-inflammatory cytokines on osteoporosis. We also highlighted the studies regarding the impact of physiological and metabolic changes in the body, which results in chronic inflammatory conditions such as ageing, ultimately triggering osteoporosis.
Collapse
Affiliation(s)
- Yogesh Saxena
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Sanjeev Routh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Arunika Mukhopadhaya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| |
Collapse
|
20
|
Cho KM, Kim YS, Lee M, Lee HY, Bae YS. Isovaleric acid ameliorates ovariectomy-induced osteoporosis by inhibiting osteoclast differentiation. J Cell Mol Med 2021; 25:4287-4297. [PMID: 33768674 PMCID: PMC8093970 DOI: 10.1111/jcmm.16482] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 03/03/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022] Open
Abstract
Osteoclasts (OCs) play important roles in bone remodelling and contribute to bone loss by increasing bone resorption activity. Excessively activated OCs cause diverse bone disorders including osteoporosis. Isovaleric acid (IVA), also known as 3-methylbutanoic acid is a 5-carbon branched-chain fatty acid (BCFA), which can be generated by bacterial fermentation of a leucine-rich diet. Here, we find that IVA suppresses differentiation of bone marrow-derived macrophages into OCs by RANKL. IVA inhibited the expression of OC-related genes. IVA-induced inhibitory effects on OC generation were attenuated by pertussis toxin but not by H89, suggesting a Gi -coupled receptor-dependent but protein kinase A-independent response. Moreover, IVA stimulates AMPK phosphorylation, and treatment with an AMPK inhibitor blocks IVA-induced inhibition of OC generation. In an ovariectomized mouse model, addition of IVA to the drinking water resulted in significant decrease of body weight gain and inhibited the expression of not only OC-related genes but also fusogenic genes in the bone tissue. IVA exposure also blocked bone destruction and OC generation in the bone tissue of ovariectomized mice. Collectively, the results demonstrate that IVA is a novel bioactive BCFA that inhibits OC differentiation, suggesting that IVA can be considered a useful material to control osteoclast-associated bone disorders, including osteoporosis.
Collapse
Affiliation(s)
- Kwang Min Cho
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Ye Seon Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Mingyu Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Ha Young Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
21
|
Insights into the mechanism of vascular endothelial cells on bone biology. Biosci Rep 2021; 41:227494. [PMID: 33403387 PMCID: PMC7816070 DOI: 10.1042/bsr20203258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/25/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022] Open
Abstract
In the skeletal system, blood vessels not only function as a conduit system for transporting gases, nutrients, metabolic waste, or cells but also provide multifunctional signal molecules regulating bone development, regeneration, and remodeling. Endothelial cells (ECs) in bone tissues, unlike in other organ tissues, are in direct contact with the pericytes of blood vessels, resulting in a closer connection with peripheral connective tissues. Close-contact ECs contribute to osteogenesis and osteoclastogenesis by secreting various cytokines in the paracrine or juxtacrine pathways. An increasing number of studies have revealed that extracellular vesicles (EVs) derived from ECs can directly regulate maturation process of osteoblasts and osteoclasts. The different pathways focus on targets at different distances, forming the basis of the intimate spatial and temporal link between bone tissue and blood vessels. Here, we provide a systematic review to elaborate on the function of ECs in bone biology and its underlying mechanisms based on three aspects: paracrine, EVs, and juxtacrine. This review proposes the possibility of a therapeutic strategy targeting blood vessels, as an adjuvant treatment for bone disorders.
Collapse
|
22
|
Van Raemdonck K, Umar S, Shahrara S. The pathogenic importance of CCL21 and CCR7 in rheumatoid arthritis. Cytokine Growth Factor Rev 2020; 55:86-93. [PMID: 32499193 PMCID: PMC10018533 DOI: 10.1016/j.cytogfr.2020.05.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 05/12/2020] [Indexed: 02/08/2023]
Abstract
Innate and adaptive immunity regulate the inflammatory and erosive phenotypes observed in rheumatoid arthritis (RA) patients. Hence, identifying novel pathways that participate in different stages of RA pathology will provide valuable insights concerning the mechanistic behavior of different joint leukocytes and the strategy to restrain their activity. Recent findings have revealed that CCL21 poses as a risk factor for RA and expression of its receptor, CCR7, on circulating monocytes is representative of the patient's disease activity score. Expression of CCR7 was found to be the hallmark of RA synovial fluid (SF) M1 macrophages (MФs) and its levels were potentiated in response to M1 mediating factors and curtailed by M2 mediators in naïve MФs. Intriguingly, although both CCR7 ligands, CCL19 and CCL21, are elevated in RA specimens, only CCL21 was predominately responsible for CCR7's pathological manifestation of RA. Unique subset of MФs differentiated in response to CCL21 stimulation, exhibited upregulation in Th17-polarizing monokines. Moreover, CCL21-activated monokines were capable of differentiating naïve T cells into joint Th17 cells, which also partook in RA osteoclastogenesis. Finally, to conserve chronic inflammation, SF CCL21 amplified RA neovascularization directly and indirectly by promoting RA FLS and MΦs to secrete proangiogenic factors, VEGF and IL-17. This review aims to shed light on the broad pathogenic impact of CCL21, linking immunostimulatory MФs with Th17 cells, while concurrently advancing RA bone destruction and neovascularization.
Collapse
Affiliation(s)
- Katrien Van Raemdonck
- Jesse Brown VA Medical Center, Chicago, IL 60612, United States; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, United States
| | - Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL 60612, United States; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, United States
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL 60612, United States; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, United States.
| |
Collapse
|
23
|
Delgado D, Garate A, Sánchez P, Bilbao AM, García Del Caño G, Salles J, Sánchez M. Biological and structural effects after intraosseous infiltrations of age-dependent platelet-rich plasma: An in vivo study. J Orthop Res 2020; 38:1931-1941. [PMID: 32129513 DOI: 10.1002/jor.24646] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 01/09/2020] [Accepted: 02/29/2020] [Indexed: 02/04/2023]
Abstract
Platelet-rich plasma (PRP) is an increasingly widespread treatment for joint pathologies. Its characteristics and administration route are variables that may influence the clinical outcome. The aim of this in vivo study was to analyze in aged rats the biological and structure effects of intraosseous infiltrations of two different types of PRP obtained from young and old donors. During 6 months intraosseous infiltrations were performed and 4 days after the last infiltration, animals were sacrificed, and bones were extracted for micro-computed tomography (micro-CT) and histological analysis. Molecular composition of the PRP of aged donors presented higher levels of proinflammatory molecules. The histological studies showed a greater cellularity of bone marrow in groups treated with PRP. Concerning micro-CT analysis, young PRP showed a better femoral bone structure according to values of percentage of trabecular bone, trabecular space, trabecular density, and subchondral bone plate volume. In summary, this study has demonstrated that intraosseous infiltrations of PRP from young donors prevent from age-related bone degeneration. This treatment could stimulate the biological processes that maintain homeostasis and bone structure and avoid osteoarticular pathologies.
Collapse
Affiliation(s)
- Diego Delgado
- Advanced Biological Therapy Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| | - Ane Garate
- Advanced Biological Therapy Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| | - Pello Sánchez
- Advanced Biological Therapy Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| | - Ane Miren Bilbao
- Arthroscopic Surgery Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| | - Gontzal García Del Caño
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Joan Salles
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Mikel Sánchez
- Advanced Biological Therapy Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain.,Arthroscopic Surgery Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| |
Collapse
|
24
|
Guo X, Xu T, Zheng J, Cui X, Li M, Wang K, Su M, Zhang H, Zheng K, Sun C, Song S, Liu H. Accumulation of synovial fluid CD19 +CD24 hiCD27 + B cells was associated with bone destruction in rheumatoid arthritis. Sci Rep 2020; 10:14386. [PMID: 32873834 PMCID: PMC7462986 DOI: 10.1038/s41598-020-71362-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 08/10/2020] [Indexed: 12/29/2022] Open
Abstract
Regulatory CD19+CD24hiCD27+ B cells were proved to be numerically decreased and functionally impaired in the peripheral blood (PB) from rheumatoid arthritis (RA), with the potential of converting into osteoclast-priming cells. However, the distribution and function of CD19+CD24hiCD27+ B cells in RA synovial fluid (SF) were unclear. In this study, we investigated whether RA SF CD19+CD24hiCD27+ B cells were increased and associated with bone destruction. We found that the proportion of RA SF CD19+CD24hiCD27+ B cells was increased significantly, and was positively correlated with swollen joint counts, tender joint counts and disease activity. CXCL12, CXCL13, CCL19 contributed to the recruitment of CD19+CD24hiCD27+ B cells in RA SF. Notably, CD19+CD24hiCD27+ B cells in the SF from RA expressed significantly more RANKL compared to OA and that in the PB from RA. Critically, RA CD19+CD24hiCD27+ B cells promoted osteoclast (OC) differentiation in vitro, and the number of OCs was higher in cultures with RA SF CD19+CD24hiCD27+ B cells than in those derived from RA PB. Collectively, these findings revealed the accumulation of CD19+CD24hiCD27+ B cells in SF and their likely contribution to joint destruction in RA. Modulating the status of CD19+CD24hiCD27+ B cells might provide novel therapeutic strategies for RA.
Collapse
Affiliation(s)
- Xiaofeng Guo
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Tingting Xu
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Jing Zheng
- Department of Hematology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, Yichang, 443000, Hubei Province, China
| | - Xiangjun Cui
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Ming Li
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Kai Wang
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Min Su
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Huifang Zhang
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Ke Zheng
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Chongling Sun
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China
| | - Shulin Song
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China.
| | - Hongjiang Liu
- Department of Rheumatology and Immunology, The People's Hospital of China Three Gorges University/The First People's Hospital of Yichang, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei Province, China.
| |
Collapse
|
25
|
Tian Y, Terkawi MA, Onodera T, Alhasan H, Matsumae G, Takahashi D, Hamasaki M, Ebata T, Aly MK, Kida H, Shimizu T, Uetsuki K, Kadoya K, Iwasaki N. Blockade of XCL1/Lymphotactin Ameliorates Severity of Periprosthetic Osteolysis Triggered by Polyethylene-Particles. Front Immunol 2020; 11:1720. [PMID: 32849609 PMCID: PMC7417302 DOI: 10.3389/fimmu.2020.01720] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/29/2020] [Indexed: 12/19/2022] Open
Abstract
Periprosthetic osteolysis induced by orthopedic implant-wear particles continues to be the leading cause of arthroplasty failure in majority of patients. Release of the wear debris results in a chronic local inflammatory response typified by the recruitment of immune cells, including macrophages. The cellular mediators derived from activated macrophages favor the osteoclast-bone resorbing activity resulting in bone loss at the site of implant and loosening of the prosthetic components. Emerging evidence suggests that chemokines and their receptors are involved in the progression of periprosthetic osteolysis associated with aseptic implant loosening. In the current study, we investigated the potential role of chemokine C-motif-ligand-1 (XCL1) in the pathogenesis of inflammatory osteolysis induced by wear particles. Expressions of XCL1 and its receptor XCR1 were evident in synovial fluids and tissues surrounding hip-implants of patients undergoing revision total hip arthroplasty. Furthermore, murine calvarial osteolysis model induced by ultra-high molecular weight polyethylene (UHMWPE) particles was used to study the role of XCL1 in the development of inflammatory osteolysis. Mice received single injection of recombinant XCL1 onto the calvariae after implantation of particles exhibited significantly greater osteolytic lesions than the control mice. In contrast, blockade of XCL1 by neutralizing antibody significantly reduced bone erosion and the number of bone-resorbing mature osteoclasts induced by UHMWPE particles. In consistence with the results, transplantation of XCL1-soaked sponge onto calvariae caused osteolytic lesions coincident with excessive infiltration of inflammatory cells and osteoclasts. These results suggested that XCL1 might be involved in the development of periprosthetic osteolysis through promoting infiltration of inflammatory cells and bone resorbing-osteoclasts. Our further results demonstrated that supplementing recombinant XCL1 to cultured human monocytes stimulated with the receptor activator of nuclear factor kappa-B ligand (RANKL) promoted osteoclastogenesis and the osteoclast-bone resorbing activity. Moreover, recombinant XCL1 promoted the expression of inflammatory and osteoclastogenic factors, including IL-6, IL-8, and RANKL in human differentiated osteoblasts. Together, these results suggested the potential role of XCL1 in the pathogenesis of periprosthetic osteolysis and aseptic loosening. Our data broaden knowledge of the pathogenesis of aseptic prosthesis loosening and highlight a novel molecular target for therapeutic intervention.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mohamad Alaa Terkawi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Global Institution for Collaborative Research and Education (GI-CoRE), Frontier Research Center for Advanced Material and Life Science, Hokkaido University, Sapporo, Japan
| | - Tomohiro Onodera
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Global Institution for Collaborative Research and Education (GI-CoRE), Frontier Research Center for Advanced Material and Life Science, Hokkaido University, Sapporo, Japan
| | - Hend Alhasan
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Gen Matsumae
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Daisuke Takahashi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masanari Hamasaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Taku Ebata
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mahmoud Khamis Aly
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroaki Kida
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tomohiro Shimizu
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Keita Uetsuki
- R&D Center, Teijin Nakashima Medical Co., Ltd., Okayama, Japan
| | - Ken Kadoya
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Global Institution for Collaborative Research and Education (GI-CoRE), Frontier Research Center for Advanced Material and Life Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
26
|
Lee S, Yoo JI, Lee YK, Park JW, Won S, Yeom J, Im JW, Lim SM, Ha YC, Koo KH. Risk of Osteoporotic Fracture in Patients with Breast Cancer: Meta-Analysis. J Bone Metab 2020; 27:27-34. [PMID: 32190606 PMCID: PMC7064363 DOI: 10.11005/jbm.2020.27.1.27] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/05/2020] [Accepted: 01/09/2020] [Indexed: 12/31/2022] Open
Abstract
Background The fracture risk induced by anti-estrogen therapy in patients with breast cancer remains controversial. The aim of this study was to perform a meta-analysis and systematic review to evaluate the risk of osteoporotic fracture in patients with breast cancer. Methods A systematic search was performed to identify studies that included any osteoporotic fracture (hip fracture and vertebral fracture) in patients breast cancer. Main outcome measures were occurrence and risk of osteoporotic fractures including hip and vertebral fractures in patients and controls. Results A systematic search yielded a total of 4 studies that included osteoporotic fracture outcomes in patients with breast cancer. Meta-analysis showed a higher risk of osteoporotic fracture in patients with breast cancer. Analysis of these 4 studies involving a total of 127,722 (23,821 cases and 103,901 controls) patients showed that the incidence of osteoporotic fractures was higher in the breast cancer group than in the control group. The pooled estimate of crude relative risk for osteoporotic fracture was 1.35 (95% confidence interval, 1.29-1.42; P<0.001). Conclusions Although studies were limited by a small number, results suggested a possible association between anti-estrogen therapy and increased risk of osteoporotic fractures in patients with breast cancer.
Collapse
Affiliation(s)
- Seeyoun Lee
- Department of Surgery, Center for Breast Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Jun-Il Yoo
- Department of Orthopaedic Surgery, Gyeongsang National University Hospital, Jinju, Korea
| | - Young-Kyun Lee
- Department of Orthopaedic Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jung-Wee Park
- Department of Orthopaedic Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Seokhyung Won
- Department of Orthopaedic Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jiung Yeom
- Department of Orthopaedic Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jin Woo Im
- Department of Orthopaedic Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Seok Min Lim
- Department of Orthopaedic Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Yong-Chan Ha
- Department of Orthopaedic Surgery, Chung-Ang University College of Medicine, Seoul, Korea
| | - Kyung-Hoi Koo
- Department of Orthopaedic Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
27
|
Elemam NM, Hannawi S, Maghazachi AA. Role of Chemokines and Chemokine Receptors in Rheumatoid Arthritis. Immunotargets Ther 2020; 9:43-56. [PMID: 32211348 PMCID: PMC7074856 DOI: 10.2147/itt.s243636] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/28/2020] [Indexed: 12/28/2022] Open
Abstract
Rheumatoid arthritis (RA) is one of the most prevalent autoimmune diseases and a prototypic inflammatory disease, affecting the small joints of the hands and feet. Chemokines and chemokine receptors play a critical role in RA pathogenesis via immune cells recruitment. Several chemokines and chemokine receptors are abundant in the peripheral blood and in the local inflamed joints of RA. Furthermore, synthetic and biologics disease modifying anti rheumatic drugs have been reported to affect chemokines expression. Thus, many studies have focused on targeting chemokines and chemokine receptors, where some have shown positive promising results. However, most of the chemokine blockers in human trials of RA treatment displayed some failures that can be attributed to several reasons in their structures and binding affinities. Nevertheless, targeting chemokines will continue to be under development, in order to improve their therapeutic potentials in RA and other autoimmune diseases. In this review we provide an up-to-date knowledge regarding the role of chemokines and chemokine receptors in RA with an emphasis on their activities on immune cells. We also discussed the effects of drugs targeting those molecules in RA. This knowledge might provide impetus for developing new therapeutic modalities to treat this chronic disease.
Collapse
Affiliation(s)
- Noha Mousaad Elemam
- College of Medicine and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Suad Hannawi
- Ministry of Health and Prevention, Department of Rheumatology, Dubai, United Arab Emirates
| | - Azzam A Maghazachi
- College of Medicine and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
28
|
Abstract
Connective tissues within the synovial joints are characterized by their dense extracellular matrix and sparse cellularity. With injury or disease, however, tissues commonly experience an influx of cells owing to proliferation and migration of endogenous mesenchymal cell populations, as well as invasion of the tissue by other cell types, including immune cells. Although this process is critical for successful wound healing, aberrant immune-mediated cell infiltration can lead to pathological inflammation of the joint. Importantly, cells of mesenchymal or haematopoietic origin use distinct modes of migration and thus might respond differently to similar biological cues and microenvironments. Furthermore, cell migration in the physiological microenvironment of musculoskeletal tissues differs considerably from migration in vitro. This Review addresses the complexities of cell migration in fibrous connective tissues from three separate but interdependent perspectives: physiology (including the cellular and extracellular factors affecting 3D cell migration), pathophysiology (cell migration in the context of synovial joint autoimmune disease and injury) and tissue engineering (cell migration in engineered biomaterials). Improved understanding of the fundamental mechanisms governing interstitial cell migration might lead to interventions that stop invasion processes that culminate in deleterious outcomes and/or that expedite migration to direct endogenous cell-mediated repair and regeneration of joint tissues.
Collapse
|
29
|
Abstract
Chemokines are a family of small proteins, subdivided by their conserved cysteine residues and common structural features. Chemokines interact with their cognate G-protein-coupled receptors to elicit downstream signals that result in cell migration, proliferation, and survival. This review presents evidence for how the various CXC and CC subfamily chemokines influence bone hemostasis by acting on osteoclasts, osteoblasts, and progenitor cells. Also discussed are the ways in which chemokines contribute to bone loss as a result of inflammatory diseases such as rheumatoid arthritis, HIV infection, and periodontal infection. Both positive and negative effects of chemokines on bone formation and bone loss are presented. In addition, the role of chemokines in altering the bone microenvironment through effects on angiogenesis and tumor invasion is discussed. Very few therapeutic agents that influence bone formation by targeting chemokines or chemokine receptors are available, although a few are currently being evaluated.
Collapse
Affiliation(s)
- Annette Gilchrist
- Department of Pharmaceutical Sciences, Midwestern University, Downers Grove, IL, USA.
| |
Collapse
|
30
|
de Oliveira Peixoto J, Savoldi IR, Ibelli AMG, Cantão ME, Jaenisch FRF, Giachetto PF, Settles ML, Zanella R, Marchesi JAP, Pandolfi JR, Coutinho LL, Ledur MC. Proximal femoral head transcriptome reveals novel candidate genes related to epiphysiolysis in broiler chickens. BMC Genomics 2019; 20:1031. [PMID: 31888477 PMCID: PMC6937697 DOI: 10.1186/s12864-019-6411-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/18/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The proximal femoral head separation (FHS) or epiphysiolysis is a prevalent disorder affecting the chicken femur epiphysis, being considered a risk factor to infection which can cause bacterial chondronecrosis with osteomyelitis in broilers. To identify the genetic mechanisms involved in epiphysiolysis, differentially expressed (DE) genes in the femur of normal and FHS-affected broilers were identified using RNA-Seq technology. Femoral growth plate (GP) samples from 35-day-old commercial male broilers were collected from 4 healthy and 4 FHS-affected broilers. Sequencing was performed using an Illumina paired-end protocol. Differentially expressed genes were obtained using the edgeR package based on the False Discovery Rate (FDR < 0.05). RESULTS Approximately 16 million reads/sample were generated with 2 × 100 bp paired-end reads. After data quality control, approximately 12 million reads/sample were mapped to the reference chicken genome (Galgal5). A total of 12,645 genes were expressed in the femur GP. Out of those, 314 were DE between groups, being 154 upregulated and 160 downregulated in FHS-affected broilers. In the functional analyses, several biological processes (BP) were overrepresented. Among them, those related to cell adhesion, extracellular matrix (ECM), bone development, blood circulation and lipid metabolism, which are more related to chicken growth, are possibly involved with the onset of FHS. On the other hand, BP associated to apoptosis or cell death and immune response, which were also found in our study, could be related to the consequence of the FHS. CONCLUSIONS Genes with potential role in the epiphysiolysis were identified through the femur head transcriptome analysis, providing a better understanding of the mechanisms that regulate bone development in fast-growing chickens. In this study, we highlighted the importance of cell adhesion and extracellular matrix related genes in triggering FHS. Furthermore, we have shown new insights on the involvement of lipidemia and immune response/inflammation with FHS in broilers. Understanding the changes in the GP transcriptome might support breeding strategies to address poultry robustness and to obtain more resilient broilers.
Collapse
Affiliation(s)
- Jane de Oliveira Peixoto
- Embrapa Suínos e Aves, Rodovia BR-153, Km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina 89715-899 Brazil
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro-Oeste, Guarapuava, Paraná, Brazil
| | - Igor Ricardo Savoldi
- Embrapa Suínos e Aves, Rodovia BR-153, Km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina 89715-899 Brazil
- Universidade do Contestado, Concórdia, Santa Catarina Brazil
- Programa de Pós-Graduação em Zootecnia, UDESC-Oeste, Chapecó, SC Brazil
| | - Adriana Mércia Guaratini Ibelli
- Embrapa Suínos e Aves, Rodovia BR-153, Km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina 89715-899 Brazil
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro-Oeste, Guarapuava, Paraná, Brazil
- Universidade do Contestado, Concórdia, Santa Catarina Brazil
| | - Maurício Egídio Cantão
- Embrapa Suínos e Aves, Rodovia BR-153, Km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina 89715-899 Brazil
| | - Fátima Regina Ferreira Jaenisch
- Embrapa Suínos e Aves, Rodovia BR-153, Km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina 89715-899 Brazil
| | | | | | - Ricardo Zanella
- Universidade de Passo Fundo, Passo Fundo, RS Brazil
- Programa de Mestrado em BioExperimentação, UPF, Passo Fundo, RS Brazil
| | - Jorge Augusto Petroli Marchesi
- Embrapa Suínos e Aves, Rodovia BR-153, Km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina 89715-899 Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP Brazil
| | - José Rodrigo Pandolfi
- Embrapa Suínos e Aves, Rodovia BR-153, Km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina 89715-899 Brazil
| | | | - Mônica Corrêa Ledur
- Embrapa Suínos e Aves, Rodovia BR-153, Km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina 89715-899 Brazil
- Programa de Pós-Graduação em Zootecnia, UDESC-Oeste, Chapecó, SC Brazil
| |
Collapse
|
31
|
George EL, Truesdell SL, Magyar AL, Saunders MM. The effects of mechanically loaded osteocytes and inflammation on bone remodeling in a bisphosphonate-induced environment. Bone 2019; 127:460-473. [PMID: 31301402 DOI: 10.1016/j.bone.2019.07.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 07/03/2019] [Accepted: 07/08/2019] [Indexed: 12/23/2022]
Abstract
Bisphosphonate-related osteonecrosis of the jaw is a disease appearing after tooth removal in patients undergoing bisphosphonate treatment for metastasizing cancers and osteoporosis. The complexity of the condition requires a multicellular model to address the net effects of two key risk factors: mechanical trauma (pathologic overload) and inflammation. In this work, a system comprised of a polydimethylsiloxane chip and mechanical loading device is used to expose bisphosphonate-treated osteocytes to mechanical trauma. Specifically, osteocytes are treated with the potent nitrogen-containing bisphosphonate, zoledronic acid, and exposed to short-term pathologic overload via substrate stretch. During bone remodeling, osteocyte apoptosis plays a role in attracting pre-osteoclasts to sites of damage; as such, lactate dehydrogenase activity, cell death and protein expression are evaluated as functions of load. Additionally, the effects of osteocyte soluble factors on osteoclast and osteoblast functional activity are quantified. Osteoclast activity and bone resorption are quantified in the presence and absence of inflammatory components, lipopolysaccharide and interferon gamma. Results suggest that inflammation associated with bacterial infection may hinder bone resorption by osteoclasts. In addition, osteocytes may respond to overload by altering expression of soluble signals that act on osteoblasts to attenuate bone formation. These findings give insight into the multicellular interactions implicated in bisphosphonate-related osteonecrosis of the jaw.
Collapse
Affiliation(s)
- Estee L George
- The University of Akron, Olson Research Center 319, 302 E. Buchtel Ave., Akron, OH 44325-0302, USA.
| | - Sharon L Truesdell
- The University of Akron, Olson Research Center 319, 302 E. Buchtel Ave., Akron, OH 44325-0302, USA.
| | - Alexandria L Magyar
- The University of Akron, Olson Research Center 319, 302 E. Buchtel Ave., Akron, OH 44325-0302, USA.
| | - Marnie M Saunders
- The University of Akron, Olson Research Center 319, 302 E. Buchtel Ave., Akron, OH 44325-0302, USA.
| |
Collapse
|
32
|
Shirakawa J, Kajikawa S, Böttcher RT, Costell M, Izu Y, Hayata T, Noda M, Ezura Y. Profilin 1 Negatively Regulates Osteoclast Migration in Postnatal Skeletal Growth, Remodeling, and Homeostasis in Mice. JBMR Plus 2019; 3:e10130. [PMID: 31346562 DOI: 10.1002/jbm4.10130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/16/2018] [Accepted: 10/21/2018] [Indexed: 01/29/2023] Open
Abstract
Profilin 1 (Pfn1), a regulator of actin polymerization, controls cell movement in a context-dependent manner. Pfn1 supports the locomotion of most adherent cells by assisting actin-filament elongation, as has been shown in skeletal progenitor cells in our previous study. However, because Pfn1 has also been known to inhibit migration of certain cells, including T cells, by suppressing branched-end elongation of actin filaments, we hypothesized that its roles in osteoclasts may be different from that of osteoblasts. By investigating the osteoclasts in culture, we first verified that Pfn1-knockdown (KD) enhances bone resorption in preosteoclastic RAW264.7 cells, despite having a comparable number and size of osteoclasts. Pfn1-KD in bone marrow cells showed similar results. Mechanistically, Pfn1-KD osteoclasts appeared more mobile than in controls. In vivo, the osteoclast-specific conditional Pfn1-deficient mice (Pfn1-cKO) by CathepsinK-Cre driver demonstrated postnatal skeletal phenotype, including dwarfism, craniofacial deformities, and long-bone metaphyseal osteolytic expansion, by 8 weeks of age. Metaphyseal and diaphyseal femurs were drastically expanded with suppressed trabecular bone mass as indicated by μCT analysis. Histologically, TRAP-positive osteoclasts were increased at endosteal metaphysis to diaphysis of Pfn1-cKO mice. The enhanced movement of Pfn1-cKO osteoclasts in culture was associated with a slight increase in cell size and podosome belt length, as well as an increase in bone-resorbing activity. Our study, for the first time, demonstrated that Pfn1 has critical roles in inhibiting osteoclast motility and bone resorption, thereby contributing to essential roles in postnatal skeletal homeostasis. Our study also provides novel insight into understanding skeletal deformities in human disorders.
Collapse
Affiliation(s)
- Jumpei Shirakawa
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Department of Oral Medicine and Stomatology School of Dental Medicine Tsurumi University Yokohama Japan
| | - Shuhei Kajikawa
- Frontier Research Unit Skeletal Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan
| | - Ralph T Böttcher
- Department of Molecular Medicine Max Planck Institute of Biochemistry Martinsried Germany
| | - Mercedes Costell
- Department of Biochemistry and Molecular Biology Faculty of Biology University of Valencia Spain
| | - Yayoi Izu
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan
| | - Tadayoshi Hayata
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Department of Molecular Pharmacology Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Science Tokyo University of Science Noda CHIBA Japan
| | - Masaki Noda
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Yokohama City Minato Red Cross Hospital Yokohama Japan.,Department of Orthopedic Surgery Tokyo Medical and Dental University Tokyo Japan
| | - Yoichi Ezura
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Frontier Research Unit Skeletal Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan
| |
Collapse
|
33
|
Shima K, Kimura K, Ishida M, Kishikawa A, Ogawa S, Qi J, Shen WR, Ohori F, Noguchi T, Marahleh A, Kitaura H. C-X-C Motif Chemokine 12 Enhances Lipopolysaccharide-Induced Osteoclastogenesis and Bone Resorption In Vivo. Calcif Tissue Int 2018; 103:431-442. [PMID: 29845409 DOI: 10.1007/s00223-018-0435-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/24/2018] [Indexed: 12/23/2022]
Abstract
C-X-C motif chemokine 12 (CXCL12) belongs to the family of CXC chemokines. Lipopolysaccharide (LPS) induces inflammation-induced osteoclastogenesis and bone resorption, and in recent years, stimulatory effects of CXCL12 on bone resorption have also been reported. In the present study, we investigated the effects of CXCL12 on LPS-induced osteoclastogenesis and bone resorption. LPS was administered with or without CXCL12 onto mouse calvariae by daily subcutaneous injection. Numbers of osteoclasts and bone resorption were significantly elevated in mice co-administered LPS and CXCL12 compared with mice administered LPS alone. Moreover, receptor activator of NF-kB ligand (RANKL) and tumor necrosis factor-α (TNF-α) mRNA levels were higher in mice co-administered LPS and CXCL12 compared with mice administered LPS alone. These in vitro results confirmed a direct stimulatory effect of CXCL12 on RANKL- and TNF-α-induced osteoclastogenesis. Furthermore, TNF-α and RANKL mRNA levels were elevated in macrophages and osteoblasts, respectively, co-treated in vitro with CXCL12 and LPS, in comparison with cells treated with LPS alone. Our results suggest that CXCL12 enhances LPS-induced osteoclastogenesis and bone resorption in vivo through a combination of increasing LPS-induced TNF-α production by macrophages, increasing RANKL production by osteoblasts, and direct enhancement of osteoclastogenesis.
Collapse
Affiliation(s)
- Kazuhiro Shima
- Division of Orthodontics and Dentofacial Orthopedics, Department of Translational Medicine, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Keisuke Kimura
- Division of Orthodontics and Dentofacial Orthopedics, Department of Translational Medicine, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Masahiko Ishida
- Division of Orthodontics and Dentofacial Orthopedics, Department of Translational Medicine, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Akiko Kishikawa
- Division of Orthodontics and Dentofacial Orthopedics, Department of Translational Medicine, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Saika Ogawa
- Division of Orthodontics and Dentofacial Orthopedics, Department of Translational Medicine, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Jiawei Qi
- Division of Orthodontics and Dentofacial Orthopedics, Department of Translational Medicine, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Wei-Ren Shen
- Division of Orthodontics and Dentofacial Orthopedics, Department of Translational Medicine, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Fumitoshi Ohori
- Division of Orthodontics and Dentofacial Orthopedics, Department of Translational Medicine, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Takahiro Noguchi
- Division of Orthodontics and Dentofacial Orthopedics, Department of Translational Medicine, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Aseel Marahleh
- Division of Orthodontics and Dentofacial Orthopedics, Department of Translational Medicine, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Hideki Kitaura
- Division of Orthodontics and Dentofacial Orthopedics, Department of Translational Medicine, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| |
Collapse
|
34
|
Integration of Gene Expression Profile Data to Screen and Verify Hub Genes Involved in Osteoarthritis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9482726. [PMID: 30186872 PMCID: PMC6112076 DOI: 10.1155/2018/9482726] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/30/2018] [Accepted: 08/05/2018] [Indexed: 12/28/2022]
Abstract
Osteoarthritis (OA) is one of the most common diseases worldwide, but the pathogenic genes and pathways are largely unclear. The aim of this study was to screen and verify hub genes involved in OA and explore potential molecular mechanisms. The expression profiles of GSE12021 and GSE55235 were downloaded from the Gene Expression Omnibus (GEO) database, which contained 39 samples, including 20 osteoarthritis synovial membranes and 19 matched normal synovial membranes. The raw data were integrated to obtain differentially expressed genes (DEGs) and were deeply analyzed by bioinformatics methods. The Gene Ontology (GO) and pathway enrichment of DEGs were performed by DAVID and Kyoto Encyclopedia of Genes and Genomes (KEGG) online analyses, respectively. The protein-protein interaction (PPI) networks of the DEGs were constructed based on data from the STRING database. The top 10 hub genes VEGFA, IL6, JUN, IL1β, MYC, IL4, PTGS2, ATF3, EGR1, and DUSP1 were identified from the PPI network. Module analysis revealed that OA was associated with significant pathways including TNF signaling pathway, cytokine-cytokine receptor interaction, and osteoclast differentiation. The qRT-PCR result showed that the expression level of IL6, VEGFA, JUN, IL-1β, and ATF3 was significantly increased in OA samples (p < 0.05), and these candidate genes could be used as potential diagnostic biomarkers and therapeutic targets of OA.
Collapse
|
35
|
Dolcino M, Pelosi A, Fiore PF, Patuzzo G, Tinazzi E, Lunardi C, Puccetti A. Long Non-Coding RNAs Play a Role in the Pathogenesis of Psoriatic Arthritis by Regulating MicroRNAs and Genes Involved in Inflammation and Metabolic Syndrome. Front Immunol 2018; 9:1533. [PMID: 30061880 PMCID: PMC6054935 DOI: 10.3389/fimmu.2018.01533] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/21/2018] [Indexed: 01/03/2023] Open
Abstract
Psoriatic arthritis (PsA) is an inflammatory arthritis, characterized by inflammation of entheses and synovium, leading to joint erosions and new bone formation. It affects 10-30% of patients with psoriasis, and has an estimated prevalence of approximately 1%. PsA is considered to be primarily an autoimmune disease, driven by autoreactive T cells directed against autoantigens present in the skin and in the joints. However, an autoinflammatory origin has recently been proposed. Long noncoding RNAs (lncRNAs) are RNAs more than 200 nucleotides in length that do not encode proteins. LncRNAs play important roles in several biological processes, including chromatin remodeling, transcription control, and post-transcriptional processing. Several studies have shown that lncRNAs are expressed in a stage-specific or lineage-specific manner in immune cells that have a role in the development, activation, and effector functions of immune cells. LncRNAs are thought to play a role in several diseases, including autoimmune disorders. Indeed, a few lncRNAs have been identified in systemic lupus erythematosus, rheumatoid arthritis, and psoriasis. Although several high-throughput studies have been performed to identify lncRNAs, their biological and pathological relevance are still unknown, and most transcriptome studies in autoimmune diseases have only assessed protein-coding transcripts. No data are currently available on lncRNAs in PsA. Therefore, by microarray analysis, we have investigated the expression profiles of more than 50,000 human lncRNAs in blood samples from PsA patients and healthy controls using Human Clariom D Affymetrix chips, suitable to detect rare and low-expressing transcripts otherwise unnoticed by common sequencing methodologies. Network analysis identified lncRNAs targeting highly connected genes in the PsA transcriptome. Such genes are involved in molecular pathways crucial for PsA pathogenesis, including immune response, glycolipid metabolism, bone remodeling, type 1 interferon, wingless related integration site, and tumor necrosis factor signaling. Selected lncRNAs were validated by RT-PCR in an expanded cohort of patients. Moreover, modulated genes belonging to meaningful pathways were validated by RT-PCR in PsA PBMCs and/or by ELISA in PsA sera. The findings indicate that lncRNAs are involved in PsA pathogenesis by regulating both microRNAs and genes and open new avenues for the identification of new biomarkers and therapeutical targets.
Collapse
Affiliation(s)
- Marzia Dolcino
- Department of Medicine, University of Verona, Verona, Italy
| | - Andrea Pelosi
- Immunology Area, Pediatric Hospital Bambino Gesù, Rome, Italy
| | | | | | - Elisa Tinazzi
- Department of Medicine, University of Verona, Verona, Italy
| | | | - Antonio Puccetti
- Department of Experimental Medicine - Section of Histology, University of Genova, Genova, Italy
| |
Collapse
|
36
|
Pemmari A, Leppänen T, Paukkeri EL, Eskelinen A, Moilanen T, Moilanen E. Gene expression in adverse reaction to metal debris around metal-on-metal arthroplasty: An RNA-Seq-based study. J Trace Elem Med Biol 2018; 48:149-156. [PMID: 29773173 DOI: 10.1016/j.jtemb.2018.03.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 01/23/2023]
Abstract
Joint replacement surgery is a standard treatment of advanced osteoarthritis (OA). Since 2000, cobalt-chromium (CoCr) metal-on-metal (MoM) implants were widely used in hip arthroplasties. Some patients developed "adverse reaction to metal debris" (ARMD) around the prosthesis, resulting in a need for revision surgery. In the present study, we addressed the pathogenesis of ARMD by genome-wide expression analysis. Pseudosynovial ARMD tissue was obtained from revision surgery of Articular Surface Replacement (ASR, DePuy, Warsaw, IN, USA) hip arthroplasties. Control tissue was 1) OA synovium from primary hip arthroplasties and 2) inflammatory pseudosynovial tissue from metal-on-plastic (MoP) implant revisions. In ARMD tissue, the expression of 1446 genes was significantly increased and that of 1881 decreased as compared to OA synovium. Genes associated with immune response, tissue development and certain leukocyte signaling pathways were enriched in the differently (FC > 2) expressed genes. The network analysis proposed PRKACB, CD2, CD52 and CD53 as the central regulators of the greatest (FC > 10) differences. When ARMD tissue was compared to MoP tissue, the expression of 16 genes was significantly higher and that of 21 lower. Many of these genes were associated with redox homeostasis, metal ion binding and transport, macrophage activation and apoptosis. Interestingly, genes central to myofibroblast (AEBP1 and DES) and osteoclast (CCL21, TREM2 and CKB) development were upregulated in the MoP tissue. In network analysis, IL8, NQO1, GSTT1 and HMOX1 were identified as potential central regulators of the changes. In conclusion, excessive amounts of CoCr debris produced by MoM hip implants induces in a group of patients a unique adverse reaction characterized with enhanced expression of genes associated with inflammation, redox homeostasis, metal ion binding and transport, macrophage activation and apoptosis.
Collapse
Affiliation(s)
- Antti Pemmari
- The Immunopharmacology Research Group, Faculty of Medicine and Life Sciences, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Tiina Leppänen
- The Immunopharmacology Research Group, Faculty of Medicine and Life Sciences, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Erja-Leena Paukkeri
- The Immunopharmacology Research Group, Faculty of Medicine and Life Sciences, University of Tampere and Tampere University Hospital, Tampere, Finland
| | | | - Teemu Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Life Sciences, University of Tampere and Tampere University Hospital, Tampere, Finland; Coxa Hospital for Joint Replacement, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Life Sciences, University of Tampere and Tampere University Hospital, Tampere, Finland.
| |
Collapse
|
37
|
Wu C, Yang P, Liu H, Xiao W, Zhao L. Increased frequency of CCR7 +CD4 + T cells from patients with primary Sjögren's syndrome: An indicator of disease activity rather than of damage severity. Cytokine 2018; 110:9-17. [PMID: 29684636 DOI: 10.1016/j.cyto.2018.04.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/26/2018] [Accepted: 04/13/2018] [Indexed: 12/11/2022]
Abstract
Expression of CCR7 on T cells has been reported to be associated with the lymphocytic migration and infiltration, which is recognized as a vital part of the pathogenesis of Primary Sjögren's syndrome (pSS). Here, we compared the expression of CCR7 on CD4+T cells between pSS patients and control groups, including healthy donors (HD) and patients with systemic lupus erythematosus (SLE) and examined correlations with disease activity and damage severity, which were evaluated by EULAR Sjögren's Syndrome Disease Activity Index (ESSDAI) and Sjogren's Syndrome Disease Damage Index (SSDDI), respectively. Peripheral blood mononuclear Cells (PBMC) were obtained from patients and controls and expressions of CCR7 were evaluated by flow cytometry. CCR7 was selectively and frequently expressed on CD4+T cells, but less on CD8+ T cells of patients with pSS. In contrast, this phenomenon was neither seen in normal subjects nor in patients with SLE. The expression level of CCR7 in the peripheral blood CD4+ T cells is closely correlated with ESSDAI, but not SSDDI. Correspondently, the chemotactic index (CI) of CD4+T cells was higher than CD8+T cells in patients with pSS. Furthermore, the CI of CD4+T cells is also higher than that of other controls, which is correlated with ESSDAI. All the findings suggested that CCR7 might play an important role in the development of pSS by mediating the migration of CD4+cells. Thus, the expression of CCR7 in CD4+ T cells is probably a useful biomarker to evaluate and monitor disease activity. CCR7 can also potentially be a novel target for the therapy of pSS.
Collapse
Affiliation(s)
- Chunling Wu
- Department of Rhematology and Immunology, The First Hospital of China Medical University, 155 Nan Jing North Street, Shenyang 110001, China
| | - Pingting Yang
- Department of Rhematology and Immunology, The First Hospital of China Medical University, 155 Nan Jing North Street, Shenyang 110001, China
| | - Haina Liu
- Department of Rhematology and Immunology, The First Hospital of China Medical University, 155 Nan Jing North Street, Shenyang 110001, China
| | - Weiguo Xiao
- Department of Rhematology and Immunology, The First Hospital of China Medical University, 155 Nan Jing North Street, Shenyang 110001, China
| | - Lijuan Zhao
- Department of Rhematology and Immunology, The First Hospital of China Medical University, 155 Nan Jing North Street, Shenyang 110001, China.
| |
Collapse
|