1
|
Gomes AC, Sousa DM, Oliveira TC, Fonseca Ó, Pinto RJ, Silvério D, Fernandes AI, Moreira AC, Silva T, Teles MJ, Pereira L, Saraiva M, Lamghari M, Gomes MS. Serum amyloid A proteins reduce bone mass during mycobacterial infections. Front Immunol 2023; 14:1168607. [PMID: 37153579 PMCID: PMC10161249 DOI: 10.3389/fimmu.2023.1168607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Osteopenia has been associated to several inflammatory conditions, including mycobacterial infections. How mycobacteria cause bone loss remains elusive, but direct bone infection may not be required. Methods Genetically engineered mice and morphometric, transcriptomic, and functional analyses were used. Additionally, inflammatory mediators and bone turnover markers were measured in the serum of healthy controls, individuals with latent tuberculosis and patients with active tuberculosis. Results and discussion We found that infection with Mycobacterium avium impacts bone turnover by decreasing bone formation and increasing bone resorption, in an IFNγ- and TNFα-dependent manner. IFNγ produced during infection enhanced macrophage TNFα secretion, which in turn increased the production of serum amyloid A (SAA) 3. Saa3 expression was upregulated in the bone of both M. avium- and M. tuberculosis-infected mice and SAA1 and 2 proteins (that share a high homology with murine SAA3 protein) were increased in the serum of patients with active tuberculosis. Furthermore, the increased SAA levels seen in active tuberculosis patients correlated with altered serum bone turnover markers. Additionally, human SAA proteins impaired bone matrix deposition and increased osteoclastogenesis in vitro. Overall, we report a novel crosstalk between the cytokine-SAA network operating in macrophages and bone homeostasis. These findings contribute to a better understanding of the mechanisms of bone loss during infection and open the way to pharmacological intervention. Additionally, our data and disclose SAA proteins as potential biomarkers of bone loss during infection by mycobacteria.
Collapse
Affiliation(s)
- Ana Cordeiro Gomes
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- *Correspondence: Ana Cordeiro Gomes,
| | - Daniela Monteiro Sousa
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | | | - Óscar Fonseca
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Mestrado em Bioquímica Clínica, Universidade de Aveiro, , Aveiro, Portugal
| | - Ricardo J. Pinto
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- IPATIMUP – Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Diogo Silvério
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Isabel Fernandes
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana C. Moreira
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Tânia Silva
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria José Teles
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- CHUSJ – Centro Hospitalar de São João, Porto, Portugal
- EPIUnit, ISPUP - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
| | - Luísa Pereira
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP – Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Margarida Saraiva
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Meriem Lamghari
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria Salomé Gomes
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
2
|
Wan F, Ma F, Wu J, Qiao X, Chen M, Li W, Ma L. Effect of Lycium barbarum Polysaccharide on Decreasing Serum Amyloid A3 Expression through Inhibiting NF- κB Activation in a Mouse Model of Diabetic Nephropathy. Anal Cell Pathol (Amst) 2022; 2022:7847135. [PMID: 35132370 PMCID: PMC8817866 DOI: 10.1155/2022/7847135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 12/06/2021] [Accepted: 01/04/2022] [Indexed: 11/25/2022] Open
Abstract
Lycium barbarum polysaccharide (LBP) as one of the main bioactive constituents of the fruit of Lycium barbarum L. (LBL.) has many pharmacological activities, but its antihyperglycemic activity is not fully understood yet. This study investigated the hypoglycemic and renal protective effects of LBP on high-fat diet/streptozotocin- (HFD/STZ-) induced diabetic nephropathy (DN) in mice. Blood glucose was assessed before and after 8-week administration of LBP, and the homeostasis model assessment-insulin resistance (HOMA-IR) index was calculated for evaluating the antidiabetic effect of LBP. Additionally, serum creatinine (sCr), blood urea nitrogen (BUN), and urine microalbumin were tested to evaluate the renal function. HE and PAS stainings were performed to evaluate the morphology and injury of the kidney. The results showed that LBP significantly reduces the glucose level and ameliorates the insulin resistance of diabetic mice. Importantly, LBP improves renal function by lowering the levels of sCr, BUN, and microalbumin in diabetic mice and relieves the injury in the renal glomeruli and tubules of the DN mice. Furthermore, LBP attenuates renal inflammation as evidenced by downregulating the mRNA levels of TNFα, IL1 β, IL6, and SAA3 in the renal cortex, as well as reducing the elevated circulating level and protein depositions of SAA3 in the kidney. In addition, our western blot results showed that NF-κB p65 nuclear translocation and the degradation of inhibitory κB-α (IκBα) occurred during the progress of inflammation, and such activated signaling was restrained by LBP. In conclusion, our findings suggest that LBP is a potential antidiabetic agent, which ameliorates the inflammation in DN through inhibiting NF-κB activation.
Collapse
Affiliation(s)
- Fengqi Wan
- Institute of Modern Physics, Chinese Academy of Sciences, No. 509 Nanchang Road, Lanzhou 730000, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou 730030, China
| | - Fulin Ma
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou 730030, China
| | - Jiaxin Wu
- School of Pharmacy, Lanzhou University, No. 99 Donggang West Road, Lanzhou 730000, China
| | - Xinyu Qiao
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou 730030, China
| | - Minxue Chen
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou 730030, China
| | - Wenjian Li
- Institute of Modern Physics, Chinese Academy of Sciences, No. 509 Nanchang Road, Lanzhou 730000, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Liang Ma
- Institute of Modern Physics, Chinese Academy of Sciences, No. 509 Nanchang Road, Lanzhou 730000, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
3
|
Saliu TP, Yazawa N, Hashimoto K, Miyata K, Kudo A, Horii M, Kamesawa M, Kumrungsee T, Yanaka N. Serum Amyloid A3 Promoter-Driven Luciferase Activity Enables Visualization of Diabetic Kidney Disease. Int J Mol Sci 2022; 23:ijms23020899. [PMID: 35055081 PMCID: PMC8779903 DOI: 10.3390/ijms23020899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/10/2022] Open
Abstract
The early detection of diabetic nephropathy (DN) in mice is necessary for the development of drugs and functional foods. The purpose of this study was to identify genes that are significantly upregulated in the early stage of DN progression and develop a novel model to non-invasively monitor disease progression within living animals using in vivo imaging technology. Streptozotocin (STZ) treatment has been widely used as a DN model; however, it also exhibits direct cytotoxicity to the kidneys. As it is important to distinguish between DN-related and STZ-induced nephropathy, in this study, we compared renal responses induced by the diabetic milieu with two types of STZ models: multiple low-dose STZ injections with a high-fat diet and two moderate-dose STZ injections to induce DN. We found 221 genes whose expression was significantly altered during DN development in both models and identified serum amyloid A3 (Saa3) as a candidate gene. Next, we applied the Saa3 promoter-driven luciferase reporter (Saa3-promoter luc mice) to these two STZ models and performed in vivo bioluminescent imaging to monitor the progression of renal pathology. In this study, to further exclude the possibility that the in vivo bioluminescence signal is related to renal cytotoxicity by STZ treatment, we injected insulin into Saa3-promoter luc mice and showed that insulin treatment could downregulate renal inflammatory responses with a decreased signal intensity of in vivo bioluminescence imaging. These results strongly suggest that Saa3 promoter activity is a potent non-invasive indicator that can be used to monitor DN progression and explore therapeutic agents and functional foods.
Collapse
|
4
|
Pan D, Qian B, Zhao D, Yao B. Nfib promotes chondrocyte proliferation and inhibits differentiation by mildly regulating Sox9 and its downstream genes. Mol Biol Rep 2021; 48:7487-7497. [PMID: 34651294 DOI: 10.1007/s11033-021-06767-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND Chondrocyte proliferation and differentiation play pivotal roles in regulating cartilage formation, endochondral bone formation, and repair. Cartilage damage and underdevelopment may cause severe joint diseases. Various transcription factors regulate cartilage development. Nuclear factor 1 B (Nfib) is a transcription factor that plays a regulatory role in various organs. However, the effect and mechanism of Nfib on the proliferation and differentiation of chondrocytes in cartilage are still largely unknown. METHODS AND RESULTS In the present study, we investigated the gene expression patterns in primary chondrocytes with Nfib overexpression or silencing by RNA sequencing (RNA-seq) technology. The results showed that Nfib overexpression significantly up-regulated genes that are related to chondrocyte proliferation and extracellular matrix (ECM) synthesis and significantly down-regulated genes related to chondrocyte differentiation and ECM degradation. However, with Nfib silencing, the genes involved in promoting chondrocyte differentiation were significantly up-regulated, whereas those involved in promoting chondrocyte proliferation were significantly down-regulated. Furthermore, quantitative real-time PCR (qRT-PCR), western blot, alcian blue staining and immunofluorescence staining assays further confirmed that Nfib potentially promotes chondrocyte proliferation and extracellular synthesis but inhibits differentiation. CONCLUSIONS The molecular mechanism of Nfib in promoting chondrocyte proliferation and inhibiting differentiation was probably achieved by stimulating Sox9 and its downstream genes. Thus, this study adds new insights regarding the underlying molecular mechanism of transcriptional regulation in cartilage.
Collapse
Affiliation(s)
- Daian Pan
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Benxin Qian
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Daqing Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China.
| | - Baojin Yao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China.
| |
Collapse
|
5
|
Kamiya S, Shimizu K, Okada A, Inoshima Y. Induction of Serum Amyloid A3 in Mouse Mammary Epithelial Cells Stimulated with Lipopolysaccharide and Lipoteichoic Acid. Animals (Basel) 2021; 11:ani11061548. [PMID: 34070499 PMCID: PMC8230092 DOI: 10.3390/ani11061548] [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: 04/07/2021] [Revised: 05/15/2021] [Accepted: 05/23/2021] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Serum amyloid A (SAA) is an acute phase protein present in mammals and birds. Based on the amino acid sequence, SAA has been classified into isoforms SAA1–4 in mice. Previously, it was reported that after the stimulation with bacterial antigens, the expression of the Saa3 mRNA was induced more strongly than that of the Saa1 mRNA in mouse epithelia, including colonic and alveolar epithelial cells, indicating that SAA3 plays a role in the local response. However, the contribution of SAA3 to the local response in mouse mammary epithelium, where mastitis occurs due to bacterial infection, has not been completely determined yet. In this study, to clarify whether mouse SAA3 has a role in the defense against bacterial infection in mouse mammary epithelium, normal murine mammary gland (NMuMG) epithelial cells were stimulated with lipopolysaccharide (LPS) and lipoteichoic acid (LTA). LPS and LTA significantly enhanced mRNA expression level of the Saa3 gene but not that of Saa1. Furthermore, LPS induced SAA3 protein expression more strongly than LTA. Our data indicate that SAA3 expression in mouse mammary epithelial cells was increased by the stimulation with bacterial antigens, suggesting that SAA3 is involved in the defense against bacterial infection in mouse mammary epithelium. Abstract In this study, to establish whether serum amyloid A (SAA) 3 plays a role in the defense against bacterial infection in mouse mammary epithelium, normal murine mammary gland (NMuMG) epithelial cells were stimulated with lipopolysaccharide (LPS) and lipoteichoic acid (LTA). LPS and LTA significantly enhanced mRNA expression level of the Saa3 gene, whereas no significant change was observed in the Saa1 mRNA level. Furthermore, LPS induced SAA3 protein expression more strongly than LTA, whereas neither LPS nor LTA significantly affected SAA1 protein expression. These data indicate that the expression of SAA3 in mouse mammary epithelial cells was increased by the stimulation with bacterial antigens. SAA3 has been reported to stimulate neutrophils in the intestinal epithelium and increase interleukin-22 expression, which induces activation of the innate immune system and production of antibacterial proteins, such as antimicrobial peptides. Therefore, collectively, these data suggest that SAA3 is involved in the defense against bacterial infection in mouse mammary epithelium.
Collapse
Affiliation(s)
- Sato Kamiya
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan; (S.K.); (K.S.); (A.O.)
| | - Kaori Shimizu
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan; (S.K.); (K.S.); (A.O.)
| | - Ayaka Okada
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan; (S.K.); (K.S.); (A.O.)
- Education and Research Center for Food Animal Health, Gifu University (GeFAH), Gifu 501-1193, Japan
| | - Yasuo Inoshima
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan; (S.K.); (K.S.); (A.O.)
- Education and Research Center for Food Animal Health, Gifu University (GeFAH), Gifu 501-1193, Japan
- Joint Graduate School of Veterinary Sciences, Gifu University, Gifu 501-1193, Japan
- The United Graduate School of Veterinary Sciences, Gifu University, Gifu 501-1193, Japan
- Correspondence: ; Tel.: +81-58-293-2863
| |
Collapse
|
6
|
Liu JH, Yue T, Luo ZW, Cao J, Yan ZQ, Jin L, Wan TF, Shuai CJ, Wang ZG, Zhou Y, Xu R, Xie H. Akkermansia muciniphila promotes type H vessel formation and bone fracture healing by reducing gut permeability and inflammation. Dis Model Mech 2020; 13:dmm043620. [PMID: 33033107 PMCID: PMC7725610 DOI: 10.1242/dmm.043620] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 09/19/2020] [Indexed: 12/16/2022] Open
Abstract
Improving revascularization is one of the major measures in fracture treatment. Moderate local inflammation triggers angiogenesis, whereas systemic inflammation hampers angiogenesis. Previous studies showed that Akkermansia muciniphila, a gut probiotic, ameliorates systemic inflammation by tightening the intestinal barrier. In this study, fractured mice intragastrically administrated with A. muciniphila were found to display better fracture healing than mice treated with vehicle. Notably, more preosteclasts positive for platelet-derived growth factor-BB (PDGF-BB) were induced by A. muciniphila at 2 weeks post fracture, coinciding with increased formation of type H vessels, a specific vessel subtype that couples angiogenesis and osteogenesis, and can be stimulated by PDGF-BB. Moreover, A. muciniphila treatment significantly reduced gut permeability and inflammation at the early stage. Dextran sulfate sodium (DSS) was used to disrupt the gut barrier to determine its role in fracture healing and whether A. muciniphila still can stimulate bone fracture healing. As expected, A. muciniphila evidently improved gut barrier, reduced inflammation and restored the impaired bone healing and angiogenesis in DSS-treated mice. Our results suggest that A. muciniphila reduces intestinal permeability and alleviates inflammation, which probably induces more PDGF-BB+ preosteoclasts and type H vessel formation in callus, thereby promoting fracture healing. This study provides the evidence for the involvement of type H vessels in fracture healing and suggests the potential of A. muciniphila as a promising strategy for bone healing.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Jiang-Hua Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Tao Yue
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhong-Wei Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jia Cao
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zi-Qi Yan
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ling Jin
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Teng-Fei Wan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ci-Jun Shuai
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, Hunan 410008, China
| | - Zheng-Guang Wang
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Yong Zhou
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Ran Xu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hui Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Organ Injury, Aging and Regenerative Medicine, Changsha, Hunan 410008 China
- Hunan Key Laboratory of Bone Joint Degeneration and Injury, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
7
|
Zhang Y, Gao H, Li H, Guo J, Ouyang B, Wang M, Xu Q, Wang J, Lv M, Guo X, Liu Q, Wei L, Ren H, Xi Y, Guo Y, Ren B, Pan S, Liu C, Ding X, Xiang H, Yu Y, Song Y, Meng L, Liu S, Wang J, Jiang Y, Shi J, Liu S, Sabir JS, Sabir MJ, Khan M, Hajrah NH, Ming-Yuen Lee S, Xu X, Yang H, Wang J, Fan G, Yang N, Liu X. The White-Spotted Bamboo Shark Genome Reveals Chromosome Rearrangements and Fast-Evolving Immune Genes of Cartilaginous Fish. iScience 2020; 23:101754. [PMID: 33251490 PMCID: PMC7677710 DOI: 10.1016/j.isci.2020.101754] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 09/17/2020] [Accepted: 10/28/2020] [Indexed: 01/27/2023] Open
Abstract
Chondrichthyan (cartilaginous fish) occupies a key phylogenetic position and is important for investigating evolutionary processes of vertebrates. However, limited whole genomes impede our in-depth knowledge of important issues such as chromosome evolution and immunity. Here, we report the chromosome-level genome of white-spotted bamboo shark. Combing it with other shark genomes, we reconstructed 16 ancestral chromosomes of bamboo shark and illustrate a dynamic chromosome rearrangement process. We found that genes on 13 fast-evolving chromosomes can be enriched in immune-related pathways. And two chromosomes contain important genes that can be used to develop single-chain antibodies, which were shown to have high affinity to human disease markers by using enzyme-linked immunosorbent assay. We also found three bone formation-related genes were lost due to chromosome rearrangements. Our study highlights the importance of chromosome rearrangements, providing resources for understanding of cartilaginous fish diversification and potential application of single-chain antibodies.
Collapse
Affiliation(s)
- Yaolei Zhang
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
- Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Lyngby, Denmark
| | - Haoyang Gao
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Hanbo Li
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Jiao Guo
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Bingjie Ouyang
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Meiniang Wang
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Qiwu Xu
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Jiahao Wang
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Meiqi Lv
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Xinyu Guo
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Qun Liu
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Likun Wei
- City University of Hongkong, Kowloon, Hongkong SAR
| | - Han Ren
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Yang Xi
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Yang Guo
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Bingzhao Ren
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Shanshan Pan
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Chuxin Liu
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Xiaoyan Ding
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Haitao Xiang
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Yingjia Yu
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Yue Song
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Lingfeng Meng
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Shanshan Liu
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Jun Wang
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Yuan Jiang
- BGI-Shenzhen, Shenzhen 518083, China
- Complete Genomics, Inc., San Jose, CA 95134, USA
| | - Jiahai Shi
- City University of Hongkong, Kowloon, Hongkong SAR
| | - Shiping Liu
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Jamal S.M. Sabir
- Department of Biological Sciences, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia
| | - Mumdooh J. Sabir
- Department of Biological Sciences, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia
| | - Muhummadh Khan
- Department of Biological Sciences, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia
| | - Nahid H. Hajrah
- Department of Biological Sciences, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, Macao, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen 518083, China
- James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen 518083, China
- James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Guangyi Fan
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, Macao, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Naibo Yang
- BGI-Shenzhen, Shenzhen 518083, China
- Complete Genomics, Inc., San Jose, CA 95134, USA
| | - Xin Liu
- BGI-Qingdao, BGI-Shenzhen, Qingdao 266555, China
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| |
Collapse
|
8
|
Zeng N, Yan ZP, Chen XY, Ni GX. Infrapatellar Fat Pad and Knee Osteoarthritis. Aging Dis 2020; 11:1317-1328. [PMID: 33014539 PMCID: PMC7505265 DOI: 10.14336/ad.2019.1116] [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: 08/02/2019] [Accepted: 11/16/2019] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis is the most prevalent arthritis typically characterized by degradation of cartilage. However, its pathogenesis is not fully understood. Currently, osteoarthritis is best considered a disease of the whole "joint organ". Infrapatellar fat pad (IFP), an adipose tissue near synovium, is now attaching importance to researchers for its inflammatory phenotype. In this narrative review, a large body of evidence has been gathered for the involvement of IFP in the development of knee osteoarthritis. Additionally, the underlying mechanisms of how IFP can be involved in this process have been proposed. However, further investigations are needed to better understand its precise role in this process and its underlying mechanism, and beyond that, to develop new strategies to slow down the degenerative process and explore an effective and timely diagnosis of the disease.
Collapse
Affiliation(s)
- Ni Zeng
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhi-Peng Yan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xin-Yuan Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guo-Xin Ni
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
- Correspondence should be addressed to: Dr. Guo-Xin Ni, School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China.
| |
Collapse
|
9
|
Structural Basis for Vital Function and Malfunction of Serum Amyloid A: an Acute-Phase Protein that Wears Hydrophobicity on Its Sleeve. Curr Atheroscler Rep 2020; 22:69. [PMID: 32968930 PMCID: PMC7511256 DOI: 10.1007/s11883-020-00888-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review addresses normal and pathologic functions of serum amyloid A (SAA), an enigmatic biomarker of inflammation and protein precursor of AA amyloidosis, a life-threatening complication of chronic inflammation. SAA is a small, highly evolutionarily conserved acute-phase protein whose plasma levels increase up to one thousand-fold in inflammation, infection, or after trauma. The advantage of this dramatic but transient increase is unclear, and the complex role of SAA in immune response is intensely investigated. This review summarizes recent advances in our understanding of the structure-function relationship of this intrinsically disordered protein, outlines its newly emerging beneficial roles in lipid transport and inflammation control, and discusses factors that critically influence its misfolding in AA amyloidosis. RECENT FINDINGS High-resolution structures of lipid-free SAA in crystals and fibrils have been determined by x-ray crystallography and electron cryo-microscopy. Low-resolution structural studies of SAA-lipid complexes, together with biochemical, cell-based, animal model, genetic, and clinical studies, have provided surprising new insights into a wide range of SAA functions. An emerging vital role of SAA is lipid encapsulation to remove cell membrane debris from sites of injury. The structural basis for this role has been proposed. The lysosomal origin of AA amyloidosis has solidified, and its molecular and cellular mechanisms have emerged. Recent studies have revealed molecular underpinnings for understanding complex functions of this Cambrian protein in lipid transport, immune response, and amyloid formation. These findings help guide the search for much-needed targeted therapies to block the protein deposition in AA amyloidosis.
Collapse
|
10
|
Huang X, Chen Z, Zhao G, Shi J, Huang G, Chen F, Wei Y, Xia J, Chen J, Wang S. Combined culture experiment of mouse bone marrow mesenchymal stem cells and bioceramic scaffolds. Exp Ther Med 2020; 20:19. [PMID: 32934684 PMCID: PMC7471870 DOI: 10.3892/etm.2020.9147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 04/24/2020] [Indexed: 01/11/2023] Open
Abstract
Articular cartilage injuries are common orthopedic conditions that severely affect the quality of life of patients. Tissue engineering can facilitate cartilage repair and the key points involve scaffolding and seed cell selection. Pre-experiments found a range of microstructures of bioceramic scaffolds suitable for chondrocyte adhesion and proliferation, and maintaining chondrocyte phenotype. Three-dimensional cultures of bone marrow mesenchymal stem cell (BMSC) scaffolds were implanted into mice. According to the shape of the bioceramic scaffolds and the implantation time in vivo, RNA sequencing was performed on the removed scaffolds to explore the molecular mechanism. The in vitro bone plate culture can induce differentiation of chondrocytes, making culture different to that produced in vitro. Implantation of scaffolds in vivo increases the expression of bone-related genes. The ceramic rod-like material was found to be superior to the disc shape, and the bone repair effect was more marked with longer implantation times. Gene Ontology analysis revealed that ‘cell chemotaxis’, ‘negative regulation of ossification’ and ‘bone development’ pathways were involved in recovery. It was further confirmed that BMSCs were suitable as seed cells for cartilage tissue engineering, and that the β-tricalcium phosphate scaffold maybe ideal as cartilage tissue engineering scaffold material. The present research provided new insights into the molecular mechanism of cartilage repair by BMSCs and bioceramic scaffolds. Bioinformatics analysis revealed that AMMECR1L-like protein, tumor necrosis factor-induced protein 2, inhibitor of nuclear factor-B kinase subunit and protein kinase C type and ‘negative regulation of ossification’ and ‘bone development’ pathways may be involved in osteoblast maturation and bone regeneration.
Collapse
Affiliation(s)
- Xin Huang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Zhenhao Chen
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Guanglei Zhao
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jingsheng Shi
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Gangyong Huang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Feiyan Chen
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Yibing Wei
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jun Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jie Chen
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Siqun Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
11
|
The host response to bacterial bone infection involves a local upregulation of several acute phase proteins. Immunobiology 2020; 225:151914. [PMID: 32098686 DOI: 10.1016/j.imbio.2020.151914] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/10/2020] [Accepted: 02/14/2020] [Indexed: 12/14/2022]
Abstract
Bone infections often become chronic and can be difficult to diagnose. In the present study, the osseous gene expression of several acute phase proteins (APPs) during osteomyelitis was investigated in a porcine model of implant associated osteomyelitis (IAO) (sampled 5, 10 and 15 days after infection) and in slaughter pigs with spontaneous hematogenous osteomyelitis, and compared to gene expression in liver tissue. Furthermore, immunohistochemical (IHC) staining of the APP complement component C3 (C3) was performed on the porcine osteomyelitis lesions together with material from human patients with chronic osteomyelitis. In the porcine bone samples a local upregulation of the expression of several APP genes, including serum amyloid A (SAA) and C3, was observed during infection. In the liver, only C-reactive protein (CRP) and Inter-Alpha-Trypsin Inhibitor Heavy Chain 4 were significantly upregulated. Serum concentrations of CRP, SAA and haptoglobin were only upregulated at day 5 in infected animals of the IAO model. This indicates a limited systemic response to osteomyelitis. Similar numbers of positive IHC stained C3 leukocytes were found in human and porcine bone samples with chronic osteomyelitis, indicating a high transcriptional value of porcine models of osteomyelitis. The local upregulation of APPs could potentially be used for diagnosing osteomyelitis.
Collapse
|
12
|
Safari A, Borhani-Haghighi A, Dianatpour M, Heydari ST, Foroughinia F, Ranjbar Omrani G. Circulating Serum Amyloid A, hs-CRP and Vitamin D Levels in Postmenopausal Osteoporosis. Galen Med J 2019; 8:e1548. [PMID: 34466525 PMCID: PMC8343903 DOI: 10.31661/gmj.v8i0.1548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/17/2019] [Accepted: 07/28/2019] [Indexed: 01/04/2023] Open
Abstract
Background: Both vitamin D and inflammation were investigated as important players in the pathogenesis of postmenopausal osteoporosis. This study compared vitamin D, inflammatory the biomarkers serum levels and their association with bone mineral density (BMD) in case and control groups to evaluate the possible immune-regulatory effect of vitamin D in this population. Materials and Methods: Participants in post-menopausal age, were categorized to 44 osteoporotic vs. 44 healthy aged-matched women according to WHO criteria. Total BMD, T- scores, Z-scores as well as fracture risk were measured in both groups, using Hologic system Dual-energy X-ray absorptiometry (DEXA). Serum 25-OH vitamin D, high sensitive CRP (hs-CRP) and serum amyloid A (SAA) were compared between groups. The association between serum biomarkers level and BMD were also investigated. The same evaluations were performed for vitamin D deficient (<20 ng/mL) and non-deficient (≥20 ng/mL) subgroups. Results: Vitamin D deficiency was higher in the osteoporotic group (32.6%) in comparison with the control group (25.6%), but the differences were not significant (P=0.47). There were no significant differences in serum levels of hs-CRP and SAA (P=0.83 and P=0.39) as well. No significant association between serum inflammatory biomarkers, vitamin D, and BMD were detected (P≥0.05). The results were the same for vitamin D deficient and non-deficient subgroups (P≥0.05). Conclusion: In the current study, the beneficial effects of vitamin D as a result of its immune-regulatory mechanisms was not reached. Larger scale studies might pave the way to define vitamin D benefits in postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Anahid Safari
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences Shiraz, Iran
| | | | - Mehdi Dianatpour
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences Shiraz, Iran
| | - Seyed Taghi Heydari
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Foroughinia
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Clinical Pharmacy Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamhossein Ranjbar Omrani
- Endocrinology & Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Correspondence to: Gholamhossein Ranjbar Omrani, Endocrinology & Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran Telephone Number: +98-713-6281569 Email Address:
| |
Collapse
|
13
|
Dieter BP, Meek RL, Anderberg RJ, Cooney SK, Bergin JL, Zhang H, Nair V, Kretzler M, Brosius FC, Tuttle KR. Serum amyloid A and Janus kinase 2 in a mouse model of diabetic kidney disease. PLoS One 2019; 14:e0211555. [PMID: 30763329 PMCID: PMC6375550 DOI: 10.1371/journal.pone.0211555] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 01/16/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Serum amyloid A (SAA), a potent inflammatory mediator, and Janus kinase 2 (JAK2), an intracellular signaling kinase, are increased by diabetes. The aims were to elucidate: 1) a JAK2-mediated pathway for increased SAA in the kidneys of diabetic mice; 2) a JAK2-SAA pathway for inflammation in podocytes. METHODS Akita diabetic mice (129S6) with podocyte JAK2 overexpression and angiotensin II infusion (4 weeks) were given a JAK1,2 inhibitor (LY03103801, 3 mg/kg/day orally for the last two weeks). Kidneys were immunostained for SAA isoform 3 (SAA3). SAA3 knockout and control mouse podocytes were exposed to advanced glycation end products (AGE) or exogenous SAA with JAK2 inhibition (Tyrphostin AG 490, 50μM). JAK2 activity (phosphorylation, Western blot, 1 hour) and mRNA for SAA3 and associated inflammatory genes (Cxcl5, Ccl2, and Ccl5) were measured by RT-PCR (20 hours). RESULTS SAA3 protein was present throughout the diabetic kidney, and podocyte JAK2 overexpression increased tubulointerstitial SAA3 compared to wild type diabetic controls, 43% versus 14% (p = 0.007); JAK1,2 inhibition attenuated the increase in SAA3 to 15% (p = 0.003). Urine albumin-to-creatinine ratio (r = 0.49, p = 0.03), mesangial index (r = 0.64, p = 0.001), and glomerulosclerosis score (r = 0.51, p = 0.02) were associated with SAA3 immunostaining scores across mouse groups. Exposing podocytes to AGE or exogenous SAA increased JAK2 activity within one hour and mRNA for associated inflammatory genes after 20 hours. JAK2 inhibition reduced SAA3 mRNA expression in podocytes exposed to AGE or SAA. SAA3 knockout podocytes had >85% lower AGE-induced inflammatory genes. CONCLUSION JAK1,2 inhibition reduced SAA and histological features of DKD in podocyte JAK2-overexpressing mice. In podocytes exposed to a diabetes-like condition, JAK2 inhibition reduced expression of SAA, while SAA knockout blocked expression of associated pro-inflammatory mediators. SAA may promote JAK2-dependent inflammation in the diabetic kidney.
Collapse
Affiliation(s)
- Brad P. Dieter
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
| | - Rick L. Meek
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
| | - Robert J. Anderberg
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
| | - Sheryl K. Cooney
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
| | - Jen L. Bergin
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
| | - Hongyu Zhang
- Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Viji Nair
- Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Matthias Kretzler
- Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Frank C. Brosius
- Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Katherine R. Tuttle
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
- Institute of Translational Health Sciences, Kidney Research Institute, Nephrology Division University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
14
|
Bindoli S, Franceschet G, Galozzi P, Zaninotto M, Camozzi V, Sfriso P. Osteoporosis in Systemic Autoinflammatory Diseases: A Case-Control Study. Front Endocrinol (Lausanne) 2019; 10:636. [PMID: 31620089 PMCID: PMC6759948 DOI: 10.3389/fendo.2019.00636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 09/03/2019] [Indexed: 01/22/2023] Open
Abstract
Objective: To assess if patients affected by systemic autoinflammatory diseases (SAIDs) present an increased risk of osteoporosis (OP). Methods: Forty adults patients referred to the Rheumatology Unit of Padova University Hospital affected by Familial Mediterranean Fever (FMF), TNF-Receptor Associated Periodic Syndrome (TRAPS), and Mevalonate Kinase Deficiency (MKD) and 40 healthy subjects were enrolled. Blood and urine samples were collected in order to define phosphocalcic metabolism, including Receptor activator of nuclear factor kappa-B ligand (RANKL) and osteoprotegerin (OPG), and among inflammatory markers serum amyloid A (SAA). Femur and lumbar dual-energy X-ray absorptiometry (DXA) scans were performed and Trabecular Bone Score (TBS) was calculated on DXA lumbar images. Results: We did not observe a statistically significant difference between Bone Mineral Density (BMD) and TBS of patients compared to controls. Also, the values of phosphocalcic metabolites in patients did not statistically differ from those in controls. However, SAA and OPG levels were significantly higher in patients compared to healthy subjects (p = 0.0244 and p = 0.0064, respectively). Conclusion: Patients of our cohort affected by FMF, TRAPS, and MKD do not present an increased risk of OP compared to the healthy controls. TBS and BMD are similar between the two groups underlining a preserved bone quality in patients. High OPG levels could suggest a protective role and a bone re-balancing action in response to an inflammatory background. Finally, it should be taken into account a modulatory role played by a pro-inflammatory cytokine such as SAA on bone homeostasis.
Collapse
Affiliation(s)
- Sara Bindoli
- Rheumatology Unit, Department of Medicine DIMED, University of Padova, Padova, Italy
| | - Giulio Franceschet
- Endocrinology Unit, Department of Medicine DIMED, University of Padova, Padova, Italy
| | - Paola Galozzi
- Rheumatology Unit, Department of Medicine DIMED, University of Padova, Padova, Italy
| | - Martina Zaninotto
- Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| | - Valentina Camozzi
- Endocrinology Unit, Department of Medicine DIMED, University of Padova, Padova, Italy
| | - Paolo Sfriso
- Rheumatology Unit, Department of Medicine DIMED, University of Padova, Padova, Italy
- *Correspondence: Paolo Sfriso
| |
Collapse
|
15
|
Lüthje FL, Skovgaard K, Jensen HE, Kruse Jensen L. Pigs are useful for the molecular study of bone inflammation and regeneration in humans. Lab Anim 2018; 52:630-640. [PMID: 29653496 DOI: 10.1177/0023677218766391] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pigs are used with increased frequency to model different kinds of orthopedic surgical conditions. In order to show the full potential of porcine models in orthopedic research, it is therefore required to examine the expression of bone regulatory genes in pigs affected by orthopedic surgery and compare it to the expression in humans and mice as mice, are one of the most applied animal species in orthopedics today. In the present study, the local molecular response to drilling of a tibial implant cavity, and the subsequent insertion of a steel implant was examined in a porcine model. Pigs were euthanized five days after drilling of the bone. The molecular response of 73 different genes was analyzed using a high-throughput quantitative polymerase chain reaction platform and compared to histopathology. Histologically, it was found that bone remodeling was initiated on day 5 after surgery and was associated with upregulation of several genes involved in bone degradation and formation ( CTSK, ACP5, IBSP, RANK, RANKL and COL1A1). Interleukin-6 and several acute-phase proteins (C3, SAA and ITIH4) were significantly upregulated, indicating their importance in the initial process of healing and osseointegration. All tested bone morphogenic proteins (BMP2, -4 and -7) including their inhibitor noggin were also significantly upregulated. Surprisingly, vascular endothelial growth factor A was not found to be regulated five days after surgery while several other vascular growth factors (ANGPT1, ANGPT2 and PTN) were upregulated. The pig was found to be a useful model for elucidation of bone regulatory genes in humans.
Collapse
Affiliation(s)
- Freja Lea Lüthje
- 1 Department of Veterinary and Animal Science, University of Copenhagen, Denmark.,2 Department of Biotechnology and Biomedicine, Technical University of Denmark, Denmark
| | - Kerstin Skovgaard
- 2 Department of Biotechnology and Biomedicine, Technical University of Denmark, Denmark
| | - Henrik Elvang Jensen
- 1 Department of Veterinary and Animal Science, University of Copenhagen, Denmark
| | - Louise Kruse Jensen
- 1 Department of Veterinary and Animal Science, University of Copenhagen, Denmark
| |
Collapse
|
16
|
Ha SW, Park J, Habib MM, Beck GR. Nano-Hydroxyapatite Stimulation of Gene Expression Requires Fgf Receptor, Phosphate Transporter, and Erk1/2 Signaling. ACS APPLIED MATERIALS & INTERFACES 2017; 9:39185-39196. [PMID: 29045789 DOI: 10.1021/acsami.7b12029] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Hydroxyapatite (HAp) is critical to health both as the main structural material of the skeleton and storage material of calcium and phosphate. Nanosized HAp (nHAp) is naturally produced by mineralizing cells during bone formation and remodeling and is the main constituent of the skeleton. As such, HAp is currently being investigated as a therapeutic biomaterial for orthopedic and dental purposes. Recent studies have suggested that extracellular nHAp can influence osteoblast lineage commitment and cell function through changes in gene expression; however, the mechanisms remain to be elucidated. Here, the cellular and molecular mechanism by which rod-shaped nHAp (10 × 100 nm) stimulates gene expression in preosteoblast bone marrow stromal cells was investigated. Electron microscopy detected a rapid and stable interaction of nHAp with the cell membrane, which correlated with a strong stimulation of the Erk1/2 signaling pathway. Results also identified the requirement of the Fgf receptor signaling and phosphate-transporters for nHAp regulated gene expression whereas a calcium-sensing receptor inhibitor had no effect. Collectively, the study uncovers novel signaling pathways and cellular events specifically stimulated by and required for the cellular response to free extracellular HAp. The results provide insight into the osteoblastic response to HAp relevant to functional mineralization and pathological calcification and could be used in the development of biomaterials for orthopedic purposes.
Collapse
Affiliation(s)
- Shin-Woo Ha
- Department of Medicine, Division of Endocrinology, Emory University , 101 Woodruff Circle, 1026 WMRB, Atlanta, Georgia 30322, United States
| | - Jonathan Park
- Department of Medicine, Division of Endocrinology, Emory University , 101 Woodruff Circle, 1026 WMRB, Atlanta, Georgia 30322, United States
| | - Mark M Habib
- The Atlanta Department of Veterans Affairs Medical Center , Decatur, Georgia 30033, United States
| | - George R Beck
- The Atlanta Department of Veterans Affairs Medical Center , Decatur, Georgia 30033, United States
- Department of Medicine, Division of Endocrinology, Emory University , 101 Woodruff Circle, 1026 WMRB, Atlanta, Georgia 30322, United States
- The Winship Cancer Institute, Emory University School of Medicine , Atlanta, Georgia 30322, United States
| |
Collapse
|
17
|
Jerez S, Araya H, Thaler R, Charlesworth MC, López-Solís R, Kalergis AM, Céspedes PF, Dudakovic A, Stein GS, van Wijnen AJ, Galindo M. Proteomic Analysis of Exosomes and Exosome-Free Conditioned Media From Human Osteosarcoma Cell Lines Reveals Secretion of Proteins Related to Tumor Progression. J Cell Biochem 2016; 118:351-360. [PMID: 27356893 DOI: 10.1002/jcb.25642] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 12/21/2022]
Abstract
Osteosarcomas are the most prevalent bone tumors in pediatric patients, but can also occur later in life. Bone tumors have the potential to metastasize to lung and occasionally other vital organs. To understand how osteosarcoma cells interact with their micro-environment to support bone tumor progression and metastasis, we analyzed secreted proteins and exosomes from three human osteosarcoma cell lines. Exosome isolation was validated by transmission electron microscopy (TEM) and immuno-blotting for characteristic biomarkers (CD63, CD9, and CD81). Exosomal and soluble proteins (less than 100 kDa) were identified by mass spectrometry analysis using nanoLC-MS/MS and classified by functional gene ontology clustering. We identified a secretome set of >3,000 proteins for both fractions, and detected proteins that are either common or unique among the three osteosarcoma cell lines. Protein ontology comparison of proteomes from exosomes and exosome-free fractions revealed differences in the enrichment of functional categories associated with different biological processes, including those related to tumor progression (i.e., angiogenesis, cell adhesion, and cell migration). The secretome characteristics of osteosarcoma cells are consistent with the pathological properties of tumor cells with metastatic potential. J. Cell. Biochem. 118: 351-360, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sofía Jerez
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Héctor Araya
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Roman Thaler
- Departments of Orthopedic Surgery & Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street S.W., MSB 3-69, Rochester, Minnesota 55905
| | | | - Remigio López-Solís
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo F Céspedes
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Amel Dudakovic
- Departments of Orthopedic Surgery & Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street S.W., MSB 3-69, Rochester, Minnesota 55905
| | - Gary S Stein
- Department of Biochemistry, University of Vermont College of Medicine, 89 Beaumont Avenue, Burlington, Vermont 05405
| | - Andre J van Wijnen
- Departments of Orthopedic Surgery & Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street S.W., MSB 3-69, Rochester, Minnesota 55905
| | - Mario Galindo
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
18
|
Sun L, Ye RD. Serum amyloid A1: Structure, function and gene polymorphism. Gene 2016; 583:48-57. [PMID: 26945629 DOI: 10.1016/j.gene.2016.02.044] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 02/24/2016] [Accepted: 02/29/2016] [Indexed: 02/07/2023]
Abstract
Inducible expression of serum amyloid A (SAA) is a hallmark of the acute-phase response, which is a conserved reaction of vertebrates to environmental challenges such as tissue injury, infection and surgery. Human SAA1 is encoded by one of the four SAA genes and is the best-characterized SAA protein. Initially known as a major precursor of amyloid A (AA), SAA1 has been found to play an important role in lipid metabolism and contributes to bacterial clearance, the regulation of inflammation and tumor pathogenesis. SAA1 has five polymorphic coding alleles (SAA1.1-SAA1.5) that encode distinct proteins with minor amino acid substitutions. Single nucleotide polymorphism (SNP) has been identified in both the coding and non-coding regions of human SAA1. Despite high levels of sequence homology among these variants, SAA1 polymorphisms have been reported as risk factors of cardiovascular diseases and several types of cancer. A recently solved crystal structure of SAA1.1 reveals a hexameric bundle with each of the SAA1 subunits assuming a 4-helix structure stabilized by the C-terminal tail. Analysis of the native SAA1.1 structure has led to the identification of a competing site for high-density lipoprotein (HDL) and heparin, thus providing the structural basis for a role of heparin and heparan sulfate in the conversion of SAA1 to AA. In this brief review, we compares human SAA1 with other forms of human and mouse SAAs, and discuss how structural and genetic studies of SAA1 have advanced our understanding of the physiological functions of the SAA proteins.
Collapse
Affiliation(s)
- Lei Sun
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Richard D Ye
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, SAR, China.
| |
Collapse
|
19
|
Thaler R, Maurizi A, Roschger P, Sturmlechner I, Khani F, Spitzer S, Rumpler M, Zwerina J, Karlic H, Dudakovic A, Klaushofer K, Teti A, Rucci N, Varga F, van Wijnen AJ. Anabolic and Antiresorptive Modulation of Bone Homeostasis by the Epigenetic Modulator Sulforaphane, a Naturally Occurring Isothiocyanate. J Biol Chem 2016; 291:6754-71. [PMID: 26757819 DOI: 10.1074/jbc.m115.678235] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Indexed: 11/06/2022] Open
Abstract
Bone degenerative pathologies like osteoporosis may be initiated by age-related shifts in anabolic and catabolic responses that control bone homeostasis. Here we show that sulforaphane (SFN), a naturally occurring isothiocyanate, promotes osteoblast differentiation by epigenetic mechanisms. SFN enhances active DNA demethylation viaTet1andTet2and promotes preosteoblast differentiation by enhancing extracellular matrix mineralization and the expression of osteoblastic markers (Runx2,Col1a1,Bglap2,Sp7,Atf4, andAlpl). SFN decreases the expression of the osteoclast activator receptor activator of nuclear factor-κB ligand (RANKL) in osteocytes and mouse calvarial explants and preferentially induces apoptosis in preosteoclastic cells via up-regulation of theTet1/Fas/Caspase 8 and Caspase 3/7 pathway. These mechanistic effects correlate with higher bone volume (∼20%) in both normal and ovariectomized mice treated with SFN for 5 weeks compared with untreated mice as determined by microcomputed tomography. This effect is due to a higher trabecular number in these mice. Importantly, no shifts in mineral density distribution are observed upon SFN treatment as measured by quantitative backscattered electron imaging. Our data indicate that the food-derived compound SFN epigenetically stimulates osteoblast activity and diminishes osteoclast bone resorption, shifting the balance of bone homeostasis and favoring bone acquisition and/or mitigation of bone resorptionin vivo Thus, SFN is a member of a new class of epigenetic compounds that could be considered for novel strategies to counteract osteoporosis.
Collapse
Affiliation(s)
- Roman Thaler
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria, Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, and
| | - Antonio Maurizi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Paul Roschger
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Ines Sturmlechner
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Farzaneh Khani
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, and
| | - Silvia Spitzer
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Monika Rumpler
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Jochen Zwerina
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Heidrun Karlic
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Amel Dudakovic
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, and
| | - Klaus Klaushofer
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Franz Varga
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria,
| | - Andre J van Wijnen
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, and
| |
Collapse
|
20
|
Choudhary S, Goetjen A, Estus T, Jacome-Galarza CE, Aguila HL, Lorenzo J, Pilbeam C. Serum Amyloid A3 Secreted by Preosteoclasts Inhibits Parathyroid Hormone-stimulated cAMP Signaling in Murine Osteoblasts. J Biol Chem 2015; 291:3882-94. [PMID: 26703472 DOI: 10.1074/jbc.m115.686576] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Indexed: 12/25/2022] Open
Abstract
Continuous parathyroid hormone (PTH) blocks its own osteogenic actions in marrow stromal cell cultures by inducing Cox2 and receptor activator of nuclear factor κB ligand (RANKL) in the osteoblastic lineage cells, which then cause the hematopoietic lineage cells to secrete an inhibitor of PTH-stimulated osteoblast differentiation. To identify this inhibitor, we used bone marrow macrophages (BMMs) and primary osteoblasts (POBs) from WT and Cox2 knock-out (KO) mice. Conditioned medium (CM) from RANKL-treated WT, but not KO, BMMs blocked PTH-stimulated cAMP production in POBs. Inhibition was reversed by pertussis toxin (PTX), which blocks Gαi/o activation. Saa3 was the most highly differentially expressed gene in a microarray comparison of RANKL-treated WT versus Cox2 KO BMMs, and RANKL induced Saa3 protein secretion only from WT BMMs. CM from RANKL-stimulated BMMs with Saa3 knockdown did not inhibit PTH-stimulated responses in POBs. SAA added to POBs inhibited PTH-stimulated cAMP responses, which was reversed by PTX. Selective agonists and antagonists of formyl peptide receptor 2 (Fpr2) suggested that Fpr2 mediated the inhibitory actions of Saa3 on osteoblasts. In BMMs committed to become osteoclasts by RANKL treatment, Saa3 expression peaked prior to appearance of multinucleated cells. Flow sorting of WT marrow revealed that Saa3 was secreted only from the RANKL-stimulated B220(-) CD3(-)CD11b(-/low) CD115(+) preosteoclast population. We conclude that Saa3 secretion from preosteoclasts, induced by RANKL in a Cox2-dependent manner, inhibits PTH-stimulated cAMP signaling and osteoblast differentiation via Gαi/o signaling. The induction of Saa3 by PTH may explain the suppression of bone formation when PTH is applied continuously and may be a new therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Shilpa Choudhary
- New England Musculoskeletal Institute, University of Connecticut Health, Farmington, Connecticut 06030 From the Departments of Medicine and
| | - Alexandra Goetjen
- New England Musculoskeletal Institute, University of Connecticut Health, Farmington, Connecticut 06030
| | - Thomas Estus
- New England Musculoskeletal Institute, University of Connecticut Health, Farmington, Connecticut 06030
| | | | | | - Joseph Lorenzo
- New England Musculoskeletal Institute, University of Connecticut Health, Farmington, Connecticut 06030 From the Departments of Medicine and
| | - Carol Pilbeam
- New England Musculoskeletal Institute, University of Connecticut Health, Farmington, Connecticut 06030 From the Departments of Medicine and
| |
Collapse
|
21
|
Kim J, Yang J, Park OJ, Kang SS, Yun CH, Han SH. Serum amyloid A inhibits osteoclast differentiation to maintain macrophage function. J Leukoc Biol 2015; 99:595-603. [DOI: 10.1189/jlb.3a0415-173r] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/19/2015] [Indexed: 01/07/2023] Open
|
22
|
Lenertz LY, Baughman CJ, Waldschmidt NV, Thaler R, van Wijnen AJ. Control of bone development by P2X and P2Y receptors expressed in mesenchymal and hematopoietic cells. Gene 2015; 570:1-7. [PMID: 26079571 DOI: 10.1016/j.gene.2015.06.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 06/05/2015] [Accepted: 06/06/2015] [Indexed: 02/06/2023]
Abstract
Bone development and homeostasis require the interplay between several cell types, including mesenchymal osteoblasts and osteocytes, as well as hematopoietic osteoclasts. Recent evidence suggests that cell proliferation, differentiation and apoptosis of both mesenchymal and hematopoietic stem cells, which are fundamental for tissue regeneration and treatment of degenerative diseases, are controlled by P2 receptors (i.e., P2X and P2Y receptors). Both types of P2 receptors are versatile transducers of diverse signals activated by extracellular nucleotides like ATP that are released in response to tissue injury, infection or shear stress. The P2X family of receptors has been shown to mediate multiple signaling events including the influx of calcium, activation of mitogen activated protein kinases (MAPKs) and induction of AP-1 family members known to regulate bone development. Support for the significance of P2X7 in regulating bone development and homeostasis has been provided by several studies focusing on animal models and single nucleotide polymorphisms. P2 receptors are functionally expressed in both bone forming osteoblasts and bone resorbing osteoclasts, while recent findings also suggest that these receptors translate mechanical stimuli in osteocytes. Their ability to respond to external nucleotide analogs renders these cell surface proteins excellent targets for skeletal regenerative therapies. This overview summarizes mechanisms by which nucleotide receptors control skeletal cells and contribute to bone tissue development remodeling and repair.
Collapse
Affiliation(s)
- Lisa Y Lenertz
- Department of Biology, St. Olaf College, Northfield, MN, USA
| | - Cory J Baughman
- Department of Biology, St. Olaf College, Northfield, MN, USA
| | | | - Roman Thaler
- Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Andre J van Wijnen
- Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|