1
|
Bi J, Mo C, Li S, Zeng J, Chai Y, Yao M, Liu Z, Yuan P, Ni J, Xu S. High concentrations of NaF aggravate periodontitis by promoting M1 polarization in macrophages. Int Immunopharmacol 2024; 140:112830. [PMID: 39096872 DOI: 10.1016/j.intimp.2024.112830] [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: 06/19/2024] [Revised: 07/15/2024] [Accepted: 07/26/2024] [Indexed: 08/05/2024]
Abstract
High-concentration fluoride treatment is commonly used to prevent dental caries in the oral cavity, and fluorine-containing protective paint is used to alleviate common root sensitivity symptoms in patients with periodontitis after periodontal treatment. Recent studies have confirmed its safe use in normal oral environments. However, whether fluoride treatment affects the progression of periodontitis in an inflammatory microenvironment remains unclear. Immunometabolism is crucial for maintaining bone regeneration and repair in periodontitis, and the precise regulation of macrophage polarisation is crucial to this process. Fluoride can influence the immune microenvironment of bone tissue by regulating immune metabolic processes. Herein, we investigated the effects of high concentrations of sodium fluoride (NaF) on periodontal tissues. We examined the expression of osteogenic and M1/M2 macrophage polarisation markers and glucose metabolism in macrophages. RNA sequencing was used to study differentially expressed genes related to M1 polarisation and glucose metabolism in treated macrophages. The results showed that NaF indirectly affects human periodontal ligament cells (hPDLCs), aggravating bone loss, tissue destruction, and submandibular lymph node drainage. Furthermore, NaF promoted glycolysis in macrophages and M1 polarisation while inhibiting osteogenic differentiation. These findings suggest that NaF has a direct effect on hPDLCs. Moreover, we found that high concentrations of NaF stimulated M1 polarisation in macrophages by promoting glycolysis. Overall, these results suggest that M1 macrophages promote the osteoclastic ability of hPDLCs and inhibit their osteogenic ability, eventually aggravating periodontitis. These findings provide important insights into the mechanism of action of NaF in periodontal tissue regeneration and reconstruction, which is critical for providing appropriate recommendations for the use of fluoride in patients with periodontitis.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Siwei Li
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yan Chai
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Mingyan Yao
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China; Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Jia Ni
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Wang Z, Chen X, Yan L, Wang W, Zheng P, Mohammadreza A, Liu Q. Antimicrobial peptides in bone regeneration: mechanism and potential. Expert Opin Biol Ther 2024; 24:285-304. [PMID: 38567503 DOI: 10.1080/14712598.2024.2337239] [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: 11/27/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Antimicrobial peptides (AMPs) are small-molecule peptides with a unique antimicrobial mechanism. Other notable biological activities of AMPs, including anti-inflammatory, angiogenesis, and bone formation effects, have recently received widespread attention. These remarkable bioactivities, combined with the unique antimicrobial mechanism of action of AMPs, have led to their increasingly important role in bone regeneration. AREAS COVERED In this review, on the one hand, we aimed to summarize information about the AMPs that are currently used for bone regeneration by reviewing published literature in the PubMed database. On the other hand, we also highlight some AMPs with potential roles in bone regeneration and their possible mechanisms of action. EXPERT OPINION The translation of AMPs to the clinic still faces many problems, but their unique antimicrobial mechanisms and other conspicuous biological activities suggest great potential. An in-depth understanding of the structure and mechanism of action of AMPs will help us to subsequently combine AMPs with different carrier systems and perform structural modifications to reduce toxicity and achieve stable release, which may be a key strategy for facilitating the translation of AMPs to the clinic.
Collapse
Affiliation(s)
- ZhiCheng Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - XiaoMan Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Liang Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - WenJie Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - PeiJia Zheng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Atashbahar Mohammadreza
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of International Education, Southern Medical University, Guangzhou, China
| | - Qi Liu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Liu F, Zhao Y, Pei Y, Lian F, Lin H. Role of the NF-kB signalling pathway in heterotopic ossification: biological and therapeutic significance. Cell Commun Signal 2024; 22:159. [PMID: 38439078 PMCID: PMC10910758 DOI: 10.1186/s12964-024-01533-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 02/13/2024] [Indexed: 03/06/2024] Open
Abstract
Heterotopic ossification (HO) is a pathological process in which ectopic bone develops in soft tissues within the skeletal system. Endochondral ossification can be divided into the following types of acquired and inherited ossification: traumatic HO (tHO) and fibrodysplasia ossificans progressiva (FOP). Nuclear transcription factor kappa B (NF-κB) signalling is essential during HO. NF-κB signalling can drive initial inflammation through interactions with the NOD-like receptor protein 3 (NLRP3) inflammasome, Sirtuin 1 (SIRT1) and AMP-activated protein kinase (AMPK). In the chondrogenesis stage, NF-κB signalling can promote chondrogenesis through interactions with mechanistic target of rapamycin (mTOR), phosphatidylinositol-3-kinase (PI3K)/AKT (protein kinase B, PKB) and other molecules, including R-spondin 2 (Rspo2) and SRY-box 9 (Sox9). NF-κB expression can modulate osteoblast differentiation by upregulating secreted protein acidic and rich in cysteine (SPARC) and interacting with mTOR signalling, bone morphogenetic protein (BMP) signalling or integrin-mediated signalling under stretch stimulation in the final osteogenic stage. In FOP, mutated ACVR1-induced NF-κB signalling exacerbates inflammation in macrophages and can promote chondrogenesis and osteogenesis in mesenchymal stem cells (MSCs) through interactions with smad signalling and mTOR signalling. This review summarizes the molecular mechanism of NF-κB signalling during HO and highlights potential therapeutics for treating HO.
Collapse
Affiliation(s)
- Fangzhou Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Yike Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Yiran Pei
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Fengyu Lian
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
4
|
Yu C, Sautchuk R, Martinez J, Eliseev RA. Mitochondrial permeability transition regulator, cyclophilin D, is transcriptionally activated by C/EBP during adipogenesis. J Biol Chem 2023; 299:105458. [PMID: 37949231 PMCID: PMC10716586 DOI: 10.1016/j.jbc.2023.105458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023] Open
Abstract
Age-related bone loss is associated with decreased bone formation, increased bone resorption, and accumulation of bone marrow fat. During aging, differentiation potential of bone marrow stromal (a.k.a. mesenchymal stem) cells (BMSCs) is shifted toward an adipogenic lineage and away from an osteogenic lineage. In aged bone tissue, we previously observed pathological opening of the mitochondrial permeability transition pore (MPTP) which leads to mitochondrial dysfunction, oxidative phosphorylation uncoupling, and cell death. Cyclophilin D (CypD) is a mitochondrial protein that facilitates opening of the MPTP. We found earlier that CypD is downregulated during osteogenesis of BMSCs leading to lower MPTP activity and, thus, protecting mitochondria from dysfunction. However, during adipogenesis, a fate alternative to osteogenesis, the regulation of mitochondrial function and CypD expression is still unclear. In this study, we observed that BMSCs have increased CypD expression and MPTP activity, activated glycolysis, and fragmented mitochondrial network during adipogenesis. Adipogenic C/EBPα acts as a transcriptional activator of expression of the CypD gene, Ppif, during this process. Inflammation-associated transcription factor NF-κB shows a synergistic effect with C/EBPα inducing Ppif expression. Overall, we demonstrated changes in mitochondrial morphology and function during adipogenesis. We also identified C/EBPα as a transcriptional activator of CypD. The synergistic activation of CypD by C/EBPα and the NF-κB p65 subunit during this process suggests a potential link between adipogenic signaling, inflammation, and MPTP gain-of-function, thus altering BMSC fate during aging.
Collapse
Affiliation(s)
- Chen Yu
- Center for Musculoskeletal Research, University of Rochester, Rochester, New York, USA; Department of Pathology, University of Rochester, Rochester, New York, USA
| | - Rubens Sautchuk
- Center for Musculoskeletal Research, University of Rochester, Rochester, New York, USA
| | - John Martinez
- Department of Biology, University of Rochester, Rochester, New York, USA
| | - Roman A Eliseev
- Center for Musculoskeletal Research, University of Rochester, Rochester, New York, USA; Department of Pathology, University of Rochester, Rochester, New York, USA; Department of Pharmacology & Physiology, University of Rochester, Rochester, New York, USA.
| |
Collapse
|
5
|
Xia C, Ou S, Yang Y, Zhang W, Wu W, Chen Q, Li W, Lu H, Wang Y, Qi Y, Xu C. ELP2-NLRP3-GSDMD/GSDME-mediated pyroptosis is induced by TNF-α in MC3T3-E1 cells during osteogenic differentiation. J Cell Mol Med 2023; 27:4093-4106. [PMID: 37830762 PMCID: PMC10746952 DOI: 10.1111/jcmm.17994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023] Open
Abstract
Tumour necrosis factor-α (TNF-α) is a cytokine involved in systemic inflammation. TNF-α slows down osteogenic differentiation, which may contribute to poor bone development in the inflammatory microenvironment. TNF-α inhibits osteogenic differentiation by activating the JAK-STAT3 pathway, of which Signal transducer and activator of transcription 3 (STAT3)-interacting protein 1 (StIP1, also known as elongator complex protein 2, ELP2) is a key protein in the JAK-STAT3 pathway. We investigated whether and how ELP2 activation mediates the TNF-α-induced pyroptosis during osteoblastic differentiation. Using in vitro cell cultures of preosteoblastic MC3T3-E1 cells, we found that TNF-α exposure causes cell pyroptosis in an inflammatory microenvironment during osteoblastic differentiation. Bioinformatics, protein docking model and co-immunoprecipitation analysis revealed an association between ELP2, STAT3 and NLRP3. Forced ELP2 expression promoted MC3T3-E1 cells pyroptosis, with an increase in the expression of STAT3, NLRP3 inflammasome, GSDMD/GSDME, osteoblast marker genes, and the activity of alkaline phosphatase. In contrast, ELP2 silencing ameliorated MC3T3-E1 cells pyroptosis, and osteogenic differentiation, especially after TNF-α stimulation. The TNF-α-induced cells pyroptosis during osteoblastic differentiation was therefore mediated by ELP2. These results suggest that ELP2 is upregulated at the pyroptosis of MC3T3-E1 cells and inhibits osteogenic differentiation in response to TNF-α through NLRP3-GSDMD/GSDME activation.
Collapse
Affiliation(s)
- Changliang Xia
- Department of OrthopaedicsThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouPeople's Republic of China
| | - Shuanji Ou
- Department of OrthopaedicsThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouPeople's Republic of China
| | - Yang Yang
- Department of OrthopaedicsThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouPeople's Republic of China
| | - Wei Zhang
- Department of OrthopaedicsThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouPeople's Republic of China
| | - Wenjiao Wu
- Department of OrthopaedicsThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouPeople's Republic of China
| | - Qi Chen
- Department of OrthopaedicsThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouPeople's Republic of China
| | - Wenjun Li
- Department of OrthopaedicsThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouPeople's Republic of China
| | - Hanyu Lu
- Department of OrthopaedicsThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouPeople's Republic of China
| | - Yeyang Wang
- Department of OrthopaedicsThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouPeople's Republic of China
| | - Yong Qi
- Department of OrthopaedicsThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouPeople's Republic of China
| | - Changpeng Xu
- Department of OrthopaedicsThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhouPeople's Republic of China
| |
Collapse
|
6
|
Xiao J, Gong X, Fu Z, Song X, Ma Q, Miao J, Cai R, Yan Z, Wang S, Li Q, Chen Y, Yang L, Bian X, Chen Y. The influence of inflammation on the characteristics of adipose-derived mesenchymal stem cells (ADMSCs) and tissue repair capability in a hepatic injury mouse model. Stem Cell Res Ther 2023; 14:334. [PMID: 37981679 PMCID: PMC10659042 DOI: 10.1186/s13287-023-03532-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/10/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are adult stem cells with self-renewal and multi-directional differentiation potential and possess the functions of immunomodulation, regulation of cell growth, and repair of damage. Over recent years, MSCs have been found to regulate the secretion of inflammatory factors and to exert regulatory effects on various lymphocytes in inflammatory states, and on the subsequent repair of tissue damage caused by inflammation. In the present study, we analyzed the effects of tissue inflammation on the characteristics of MSCs. METHODS Human fat derived from the infrapatellar fat pad (IPFP) of knees with differing degrees of inflammation was extracted from specimens derived from total knee arthroplasties. HE and immunohistochemical staining was performed to directly observe the evidence and degree of inflammation in human infrapatellar fat pad tissue in order to classify MSCs cells, by their origin, into highly inflamed and lowly inflamed groups, and to study the effect of tissue inflammation on cell acquisition rates via cellular counting data. Flow cytometry assays were performed to investigate the effect of tissue inflammation on MSC surface marker expression. Trilineage differentiation, including osteogenesis, adipogenesis, and chondrogenesis, was performed to assess the effect of tissue inflammation on the ability of MSCs to undergo directed differentiation. The effect of tissue inflammation on the ability of MSCs to proliferate was investigated via clone formation studies. RNA-sequencing was performed to evaluate the transcriptomes of MSCs derived from different areas of inflammation. The effect of tissue inflammation on tissue repair capacity and safety of MSCs was investigated via a murine model of acute liver injury. RESULTS The results of cell count data indicate that a high degree of tissue inflammation significantly decreases the acquisition rate of MSCs, and the proportion of CD34+ and CD146+ cells. The results of our trilineage differentiation assay show that a higher degree of inflammation decreases osteogenic differentiation and enhances adipogenic and chondrogenic differentiation of MSCs. However, these differences were not statistically significant. Clone formation assays indicate that the degree of tissue inflammation at the MSC source does not significantly affect the proliferative capacity of MSCs. The transcriptomes of MSCs remain relatively stable in fat pad tissues derived from both highly and lowly inflamed samples. The results of acute liver injury investigations in mice indicate that MSCs of high and low inflammatory tissue origin have no significant difference in their tissue repair capability. CONCLUSIONS High tissue inflammation at the source of MSCs reduces the acquisition rate of MSCs and the percentage of CD34+ and CD146+ cells acquisition. However, source tissue inflammation may not significantly affect trilineage differentiation potential and proliferative capacity of MSCs. Also, MSCs obtained from differing source degrees of inflammation retain stable and similar transcriptomic profile and are both safe and efficacious for tissue repair/regeneration without detectable differences.
Collapse
Affiliation(s)
- Jingfang Xiao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Xiaoyuan Gong
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Zhenlan Fu
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Xiongbo Song
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Qinghua Ma
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Jingya Miao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Ruili Cai
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Zexuan Yan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Shuai Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Qian Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yaokai Chen
- Biobank and Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, People's Republic of China
| | - Liu Yang
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China.
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China.
| | - Yemiao Chen
- Biobank and Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, People's Republic of China.
| |
Collapse
|
7
|
Thompson AL, Grenald SA, Ciccone HA, Mohty D, Smith AF, Coleman DL, Bahramnejad E, De Leon E, Kasper-Conella L, Uhrlab JL, Margolis DS, Salvemini D, Largent-Milnes TM, Vanderah TW. Morphine-induced osteolysis and hypersensitivity is mediated through toll-like receptor-4 in a murine model of metastatic breast cancer. Pain 2023; 164:2463-2476. [PMID: 37326644 PMCID: PMC10578422 DOI: 10.1097/j.pain.0000000000002953] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/28/2023] [Accepted: 04/18/2023] [Indexed: 06/17/2023]
Abstract
ABSTRACT The propensity for breast cancer to metastasize to bone is coupled to the most common complaint among breast cancer patients: bone pain. Classically, this type of pain is treated using escalating doses of opioids, which lack long-term efficacy due to analgesic tolerance, opioid-induced hypersensitivity, and have recently been linked to enhanced bone loss. To date, the molecular mechanisms underlying these adverse effects have not been fully explored. Using an immunocompetent murine model of metastatic breast cancer, we demonstrated that sustained morphine infusion induced a significant increase in osteolysis and hypersensitivity within the ipsilateral femur through the activation of toll-like receptor-4 (TLR4). Pharmacological blockade with TAK242 (resatorvid) as well as the use of a TLR4 genetic knockout ameliorated the chronic morphine-induced osteolysis and hypersensitivity. Genetic MOR knockout did not mitigate chronic morphine hypersensitivity or bone loss. In vitro studies using RAW264.7 murine macrophages precursor cells demonstrated morphine-enhanced osteoclastogenesis that was inhibited by the TLR4 antagonist. Together, these data indicate that morphine induces osteolysis and hypersensitivity that are mediated, in part, through a TLR4 receptor mechanism.
Collapse
Affiliation(s)
- Austen L. Thompson
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Shaness A. Grenald
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Haley A. Ciccone
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Dieter Mohty
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Angela F. Smith
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Deziree L. Coleman
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Erfan Bahramnejad
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Erick De Leon
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | - Logan Kasper-Conella
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | | | - David S. Margolis
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
- Orthopaedic Surgery, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Daniela Salvemini
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Tally M. Largent-Milnes
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
- Comprehensive Pain and Addiction Center, University of Arizona, Tucson, AZ, United States
| | - Todd W. Vanderah
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
- Comprehensive Pain and Addiction Center, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
8
|
Boyce BF, Li J, Yao Z, Xing L. Nuclear Factor-Kappa B Regulation of Osteoclastogenesis and Osteoblastogenesis. Endocrinol Metab (Seoul) 2023; 38:504-521. [PMID: 37749800 PMCID: PMC10613774 DOI: 10.3803/enm.2023.501] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 09/27/2023] Open
Abstract
Maintenance of skeletal integrity requires the coordinated activity of multinucleated bone-resorbing osteoclasts and bone-forming osteoblasts. Osteoclasts form resorption lacunae on bone surfaces in response to cytokines by fusion of precursor cells. Osteoblasts are derived from mesenchymal precursors and lay down new bone in resorption lacunae during bone remodeling. Nuclear factorkappa B (NF-κB) signaling regulates osteoclast and osteoblast formation and is activated in osteoclast precursors in response to the essential osteoclastogenic cytokine, receptor activator of NF-κB ligand (RANKL), which can also control osteoblast formation through RANK-RANKL reverse signaling in osteoblast precursors. RANKL and some pro-inflammatory cytokines, including tumor necrosis factor (TNF), activate NF-κB signaling to positively regulate osteoclast formation and functions. However, these cytokines also limit osteoclast and osteoblast formation through NF-κB signaling molecules, including TNF receptor-associated factors (TRAFs). TRAF6 mediates RANKL-induced osteoclast formation through canonical NF-κB signaling. In contrast, TRAF3 limits RANKL- and TNF-induced osteoclast formation, and it restricts transforming growth factor β (TGFβ)-induced inhibition of osteoblast formation in young and adult mice. During aging, neutrophils expressing TGFβ and C-C chemokine receptor type 5 (CCR5) increase in bone marrow of mice in response to increased NF-κB-induced CC motif chemokine ligand 5 (CCL5) expression by mesenchymal progenitor cells and injection of these neutrophils into young mice decreased bone mass. TGFβ causes degradation of TRAF3, resulting in decreased glycogen synthase kinase-3β/β-catenin-mediated osteoblast formation and age-related osteoporosis in mice. The CCR5 inhibitor, maraviroc, prevented accumulation of TGFβ+/CCR5+ neutrophils in bone marrow and increased bone mass by inhibiting bone resorption and increasing bone formation in aged mice. This paper updates current understanding of how NF-κB signaling is involved in the positive and negative regulation of cytokine-mediated osteoclast and osteoblast formation and activation with a focus on the role of TRAF3 signaling, which can be targeted therapeutically to enhance bone mass.
Collapse
Affiliation(s)
- Brendan F. Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Jinbo Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
9
|
Teixeira AOM, Rodrigues-Junior V, Rodrigues BR, Souza DM, Gaia LFP, Rodrigues DBR. Comparative Analysis of TNF-alpha, TNF-R1, and TNF-R2 in Patients with Low-impact Fractures Due to Osteoporosis. Rev Bras Ortop 2023; 58:495-499. [PMID: 37396087 PMCID: PMC10310416 DOI: 10.1055/s-0042-1757963] [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: 11/28/2021] [Accepted: 09/12/2022] [Indexed: 07/04/2023] Open
Abstract
Objective To analyze the serum levels of TNF-alpha and its TNF-R1 and TNF-R2 receptors in the blood of patients with low-impact fractures due to osteoporosis, comparing between genders and with healthy patients. Methods The present study was conducted with a blood sample of 62 patients, divided into patients with osteoporosis and healthy patients. The results were obtained using the ELISA method. Cytokine concentrations were determined based on the absorbance values obtained. Results Serum TNF-alpha levels were undetectable in female patients, while in males they were found only in one patient, with no significant difference. Similar results were found in the analyses of TNF-R1 and TNF-R2 levels, a significant increase in levels of TNF-alpha receptors in the groups of patients with osteoporosis compared with the control group in both sexes. There was no significant difference between the sexes in the dosage of both receptors within the group with osteoporosis. There was also a positive and significant correlation in the levels of TNF-R1 and TNF-R2 only in women. Conclusion The significant increase in TNF-R1 and TNF-R2 levels in women with osteoporosis suggest that the release and expression of these receptors may be contributing differently to the development of osteoporosis in men and women.
Collapse
|
10
|
Han Y, Zhou Y, Li H, Gong Z, Liu Z, Wang H, Wang B, Ye X, Liu Y. Identification of diagnostic mRNA biomarkers in whole blood for ankylosing spondylitis using WGCNA and machine learning feature selection. Front Immunol 2022; 13:956027. [PMID: 36172367 PMCID: PMC9510835 DOI: 10.3389/fimmu.2022.956027] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/24/2022] [Indexed: 11/28/2022] Open
Abstract
Ankylosing spondylitis (AS) is a common inflammatory spondyloarthritis affecting the spine and sacroiliac joint that finally results in sclerosis of the axial skeleton. Aside from human leukocyte antigen B27, transcriptomic biomarkers in blood for AS diagnosis still remain unknown. Hence, this study aimed to investigate credible AS-specific mRNA biomarkers from the whole blood of AS patients by analyzing an mRNA expression profile (GSE73754) downloaded Gene Expression Omnibus, which includes AS and healthy control blood samples. Weighted gene co-expression network analysis was performed and revealed three mRNA modules associated with AS. By performing gene set enrichment analysis, the functional annotations of these modules revealed immune biological processes that occur in AS. Several feature mRNAs were identified by analyzing the hubs of the protein-protein interaction network, which was based on the intersection between differentially expressed mRNAs and mRNA modules. A machine learning-based feature selection method, SVM-RFE, was used to further screen out 13 key feature mRNAs. After verifying by qPCR, IL17RA, Sqstm1, Picalm, Eif4e, Srrt, Lrrfip1, Synj1 and Cxcr6 were found to be significant for AS diagnosis. Among them, Cxcr6, IL17RA and Lrrfip1 were correlated with severity of AS symptoms. In conclusion, our findings provide a framework for identifying the key mRNAs in whole blood of AS that is conducive for the development of novel diagnostic markers for AS.
Collapse
Affiliation(s)
- Yaguang Han
- Department of Orthopaedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yiqin Zhou
- Department of Orthopaedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Radiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haobo Li
- Department of Orthopaedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhenyu Gong
- Department of Neurosurgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Ziye Liu
- Department of Orthopaedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Huan Wang
- Department of Orthopaedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bo Wang
- Department of Orthopaedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Bo Wang, ; Xiaojian Ye, ; Yi Liu,
| | - Xiaojian Ye
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Bo Wang, ; Xiaojian Ye, ; Yi Liu,
| | - Yi Liu
- Department of Orthopaedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Bo Wang, ; Xiaojian Ye, ; Yi Liu,
| |
Collapse
|
11
|
Mao R, Zong N, Hu Y, Chen Y, Xu Y. Neuronal Death Mechanisms and Therapeutic Strategy in Ischemic Stroke. Neurosci Bull 2022; 38:1229-1247. [PMID: 35513682 PMCID: PMC9554175 DOI: 10.1007/s12264-022-00859-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/18/2022] [Indexed: 12/17/2022] Open
Abstract
Ischemic stroke caused by intracranial vascular occlusion has become increasingly prevalent with considerable mortality and disability, which gravely burdens the global economy. Current relatively effective clinical treatments are limited to intravenous alteplase and thrombectomy. Even so, patients still benefit little due to the short therapeutic window and the risk of ischemia/reperfusion injury. It is therefore urgent to figure out the neuronal death mechanisms following ischemic stroke in order to develop new neuroprotective strategies. Regarding the pathogenesis, multiple pathological events trigger the activation of cell death pathways. Particular attention should be devoted to excitotoxicity, oxidative stress, and inflammatory responses. Thus, in this article, we first review the principal mechanisms underlying neuronal death mediated by these significant events, such as intrinsic and extrinsic apoptosis, ferroptosis, parthanatos, pyroptosis, necroptosis, and autophagic cell death. Then, we further discuss the possibility of interventions targeting these pathological events and summarize the present pharmacological achievements.
Collapse
Affiliation(s)
- Rui Mao
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Ningning Zong
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Yujie Hu
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Ying Chen
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Yun Xu
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China.
- The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, 210008, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, China.
- Nanjing Neurology Clinic Medical Center, Nanjing, 210008, China.
| |
Collapse
|
12
|
Shen G, Liu X, Lei W, Duan R, Yao Z. Plumbagin is a NF-κB-inducing kinase inhibitor with dual anabolic and antiresorptive effects that prevents menopausal-related osteoporosis in mice. J Biol Chem 2022; 298:101767. [PMID: 35235833 PMCID: PMC8958545 DOI: 10.1016/j.jbc.2022.101767] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/03/2022] Open
Abstract
Osteoporosis is caused by enhanced bone resorption and relatively reduced bone formation. There is an unmet need to develop new agents with both antiresorptive and anabolic effects to treat osteoporosis, although drugs with either effect alone are available. A small molecular compound, plumbagin, was reported to inhibit receptor activator of nuclear factor kappa-B ligand-induced osteoclast (OC) differentiation by inhibiting IκBα phosphorylation-mediated canonical NF-κB activation. However, the key transcriptional factor RelA/p65 in canonical NF-κB pathway functions to promote OC precursor survival but not terminal OC differentiation. Here, we found that plumbagin inhibited the activity of NF-κB inducing kinase, the key molecule that controls noncanonical NF-κB signaling, in an ATP/ADP-based kinase assay. Consistent with this, plumbagin inhibited processing of NF-κB2 p100 to p52 in the progenitor cells of both OCs and osteoblasts (OBs). Interestingly, plumbagin not only inhibited OC but also stimulated OB differentiation in vitro. Importantly, plumbagin prevented trabecular bone loss in ovariectomized mice. This was associated with decreased OC surfaces on trabecular surface and increased parameters of OBs, including OB surface on trabecular surface, bone formation rate, and level of serum osteocalcin, compared to vehicle-treated mice. In summary, we conclude that plumbagin is a NF-κB-inducing kinase inhibitor with dual anabolic and antiresorptive effects on bone and could represent a new class of agent for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Gengyang Shen
- Department of Pathology and Laboratory Medicine, and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Xin Liu
- Department of Pathology and Laboratory Medicine, and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Wei Lei
- Department of Pathology and Laboratory Medicine, and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Rong Duan
- Department of Pathology and Laboratory Medicine, and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA.
| |
Collapse
|
13
|
Pharmacogenomics of Anti-TNF Treatment Response Marks a New Era of Tailored Rheumatoid Arthritis Therapy. Int J Mol Sci 2022; 23:ijms23042366. [PMID: 35216481 PMCID: PMC8879844 DOI: 10.3390/ijms23042366] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/19/2022] [Accepted: 02/19/2022] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is the most commonly occurring chronic inflammatory arthritis, the exact mechanism of which is not fully understood. Tumor Necrosis Factor (TNF)-targeting drugs has been shown to exert high effectiveness for RA, which indicates the key importance of this cytokine in this disease. Nevertheless, the response to TNF inhibitors varies, and approximately one third of RA patients are non-responders, which is explained by the influence of genetic factors. Knowledge in the field of pharmacogenomics of anti-TNF drugs is growing, but has not been applied in the clinical practice so far. Different genome-wide association studies identified a few single nucleotide polymorphisms associated with anti-TNF treatment response, which largely map genes involved in T cell function. Studies of the gene expression profile of RA patients have also indicated specific gene signatures that may be useful to develop novel prognostic tools. In this article, we discuss the significance of TNF in RA and present the current knowledge in pharmacogenomics related to anti-TNF treatment response.
Collapse
|
14
|
Kitaura H, Marahleh A, Ohori F, Noguchi T, Nara Y, Pramusita A, Kinjo R, Ma J, Kanou K, Mizoguchi I. Role of the Interaction of Tumor Necrosis Factor-α and Tumor Necrosis Factor Receptors 1 and 2 in Bone-Related Cells. Int J Mol Sci 2022; 23:ijms23031481. [PMID: 35163403 PMCID: PMC8835906 DOI: 10.3390/ijms23031481] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023] Open
Abstract
Tumor necrosis factor-α (TNF-α) is a pleiotropic cytokine expressed by macrophages, monocytes, and T cells, and its expression is triggered by the immune system in response to pathogens and their products, such as endotoxins. TNF-α plays an important role in host defense by inducing inflammatory reactions such as phagocytes and cytocidal systems activation. TNF-α also plays an important role in bone metabolism and is associated with inflammatory bone diseases. TNF-α binds to two cell surface receptors, the 55kDa TNF receptor-1 (TNFR1) and the 75kDa TNF receptor-2 (TNFR2). Bone is in a constant state of turnover; it is continuously degraded and built via the process of bone remodeling, which results from the regulated balance between bone-resorbing osteoclasts, bone-forming osteoblasts, and the mechanosensory cell type osteocytes. Precise interactions between these cells maintain skeletal homeostasis. Studies have shown that TNF-α affects bone-related cells via TNFRs. Signaling through either receptor results in different outcomes in different cell types as well as in the same cell type. This review summarizes and discusses current research on the TNF-α and TNFR interaction and its role in bone-related cells.
Collapse
|
15
|
Zakaria AA, Noor MHM, Ahmad H, Hassim HA, Mazlan M, Latip MQA. A Review on Therapeutic Effects of Labisia pumila on Female Reproductive Diseases. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9928199. [PMID: 34568497 PMCID: PMC8460362 DOI: 10.1155/2021/9928199] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/30/2021] [Indexed: 11/18/2022]
Abstract
The Labisia pumila (LP) is a traditional plant that is locally known as Kacip Fatimah, Selusuh Fatimah, or Pokok Ringgang by the Malaysian indigenous people. It is believed to facilitate their childbirth, treating their postchild birth and menstrual irregularities. The water extract of LP has shown to contain bioactive compounds such as flavonoids, ascorbic acid, β-carotene, anthocyanin, and phenolic acid, which contribute extensive antioxidant, anti-inflammatory, antimicrobial, and antifungal. The LP ethanolic extract exhibits significant estrogenic effects on human endomentrial adenocarcinoma cell in estrogen-free basal medium and promoting an increase in secretion of alkaline phosphate. Water based has been used for many generations, and studies had reported that it could displace in binding the antibodies and increase the estradiol production making it similar to esterone and estradiol hormone. LP extract poses a potential and beneficial aspect in medical and cosmeceutical applications. This is mainly due to its phytoestrogen properties of the LP. However, there is a specific functionality in the application of LP extract, due to specific functional group in phytoconstituent of LP. Apart from that, the extraction solvent is important in preparing the LP extract as it poses some significant and mild side effects towards consuming the LP extracts. The current situation of women reproductive disease such as postmenopausal syndrome and polycystic ovary syndrome is increasing. Thus, it is important to find ways in alternative treatment for women reproductive disease that is less costly and low side effects. In conclusion, these studies proven that LP has the potential to be an alternative way in treating female reproductive related diseases such as in postmenopausal and polysystic ovarian syndrome women.
Collapse
Affiliation(s)
- Alif Aiman Zakaria
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Mohd Hezmee Mohd Noor
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Hafandi Ahmad
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Hasliza Abu Hassim
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Mazlina Mazlan
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Mohd Qayyum Ab Latip
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
16
|
Zhao J, Liu R, Zhu J, Chen S, Xu L. Human gingiva-derived mesenchymal stem cells promote osteogenic differentiation through their immunosuppressive function. J Oral Biosci 2021:S1349-0079(21)00092-X. [PMID: 34284117 DOI: 10.1016/j.job.2021.07.003] [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: 05/31/2021] [Revised: 06/29/2021] [Accepted: 07/07/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Human gingiva-derived mesenchymal stem cells (GMSCs) have emerged as a new MSC population exhibiting robust immune regulatory functions, multipotent differentiation potential, and regenerative ability. However, the effects of GMSCs on T cells remain unexplored. Herein, we aimed to evaluate whether GMSCs promote osteogenic differentiation by regulating immune cells. METHODS The GMSC phenotype was confirmed using the colony-forming assay, immunophenotyping, Oil red O staining, and Alizarin red staining. mRNA expression levels of proinflammatory molecules (interleukin-1β [IL-1β] and tumor necrosis factor-α [TNF-α]) and anti-inflammatory factors (IL-10) were measured by quantitative reverse-transcription PCR (qRT-PCR). Then, MC3T3-E1 cells were treated with the collected co-culture supernatant, followed by alkaline phosphatase (ALP) and immunofluorescence staining to evaluate osteogenic differentiation of MC3T3-E1 cells. qRT-PCR and western blotting were employed to analyze the expression levels of osteogenic differentiation proteins, including collagen type I (COL-1), ALP, osteopontin (OPN), and runt-related transcription factor 2 (RUNX2). RESULTS GMSCs were successfully isolated and identified. We observed that GMSCs suppressed the activated T-cell function by downregulating IL-1β and TNF-α and upregulating IL-10. Simultaneously, the expression levels of osteogenesis-related genes (COL-1, ALP, OPN, and RUNX2) were markedly lower in the co-culture supernatant and Jurkat T cell supernatant groups than those in the normal culture medium group; however, expression levels were significantly increased in the co-culture supernatant group when compared with the Jurkat T cell supernatant group. CONCLUSION Our findings indicate that GMSCs could promote the osteogenic differentiation of MC3T3-E1 cells by inhibiting the biological activity of activated T cells.
Collapse
Affiliation(s)
- Jing Zhao
- School of Stomatology of Qingdao University, Qingdao, China
| | - Rui Liu
- Department of Clinical Medicine of Qingdao University, Qingdao, China
| | - Jing Zhu
- Affiliated Hospital of Weifang Medical College, Weifang, China
| | - Shulan Chen
- School of Stomatology of Qingdao University, Qingdao, China.
| | - Ling Xu
- Department of Prosthodontics, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China.
| |
Collapse
|
17
|
Wan Q, Qin W, Ma Y, Shen M, Li J, Zhang Z, Chen J, Tay FR, Niu L, Jiao K. Crosstalk between Bone and Nerves within Bone. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003390. [PMID: 33854888 PMCID: PMC8025013 DOI: 10.1002/advs.202003390] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/29/2020] [Indexed: 05/11/2023]
Abstract
For the past two decades, the function of intrabony nerves on bone has been a subject of intense research, while the function of bone on intrabony nerves is still hidden in the corner. In the present review, the possible crosstalk between bone and intrabony peripheral nerves will be comprehensively analyzed. Peripheral nerves participate in bone development and repair via a host of signals generated through the secretion of neurotransmitters, neuropeptides, axon guidance factors and neurotrophins, with additional contribution from nerve-resident cells. In return, bone contributes to this microenvironmental rendezvous by housing the nerves within its internal milieu to provide mechanical support and a protective shelf. A large ensemble of chemical, mechanical, and electrical cues works in harmony with bone marrow stromal cells in the regulation of intrabony nerves. The crosstalk between bone and nerves is not limited to the physiological state, but also involved in various bone diseases including osteoporosis, osteoarthritis, heterotopic ossification, psychological stress-related bone abnormalities, and bone related tumors. This crosstalk may be harnessed in the design of tissue engineering scaffolds for repair of bone defects or be targeted for treatment of diseases related to bone and peripheral nerves.
Collapse
Affiliation(s)
- Qian‐Qian Wan
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Wen‐Pin Qin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Yu‐Xuan Ma
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Min‐Juan Shen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Jing Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Zi‐Bin Zhang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Ji‐Hua Chen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Franklin R. Tay
- College of Graduate StudiesAugusta UniversityAugustaGA30912USA
| | - Li‐Na Niu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Kai Jiao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| |
Collapse
|
18
|
Chen S, Liang H, Ji Y, Kou H, Zhang C, Shang G, Shang C, Song Z, Yang L, Liu L, Wang Y, Liu H. Curcumin Modulates the Crosstalk Between Macrophages and Bone Mesenchymal Stem Cells to Ameliorate Osteogenesis. Front Cell Dev Biol 2021; 9:634650. [PMID: 33634135 PMCID: PMC7900185 DOI: 10.3389/fcell.2021.634650] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/04/2021] [Indexed: 01/08/2023] Open
Abstract
Bone healing is thought to be influenced by the cross-talk between bone forming and immune cells. In particular, macrophages play a crucial role in the regulation of osteogenesis. Curcumin, the major bioactive polyphenolic ingredient of turmeric, has been shown to regulate inflammatory response and osteogenic activities. However, whether curcumin could regulate macrophage polarization and subsequently influence osteogenesis remain to be elucidated. In this study, the potential immunomodulatory capability of curcumin on inflammatory response and phenotype switch of macrophages and the subsequent impact on osteogenic differentiation of MSCs are investigated. We demonstrated that curcumin exhibited significant anti-inflammatory effect by polarizing the macrophages toward anti-inflammatory phenotype, with increased expression of IL-4, IL-10, and CD206, and decreased expression of IL-1β, TNF-α, CCR7, and iNOS. In addition, curcumin could improve the osteo-immune microenvironment via promoting osteogenesis-related regenerative cytokine BMP-2 and TGF-β production. Moreover, the co-cultured test of macrophages and BMSCs showed that curcumin-modulated macrophages conditioned medium could promote osteogenic differentiation of BMSCs with increased gene (ALP, Runx-2, OCN, and OPN) and protein (Runx-2 and OCN) expression levels, enhanced ALP activity, and obvious formation of mineralized nodules. Taken together, with the interaction between curcumin-conditioned macrophage and curcumin-stimulated BMSCs, curcumin could remarkably enhance the osteogenic differentiation of BMSCs in LPS-activated inflammatory macrophage-BMSCs coculture system.
Collapse
Affiliation(s)
- Songfeng Chen
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hang Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanhui Ji
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongwei Kou
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guowei Shang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunfeng Shang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zongmian Song
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Yang
- Department of Paediatrics, The Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Lei Liu
- Department of Paediatrics, The Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yongkui Wang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongjian Liu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Ragipoglu D, Dudeck A, Haffner-Luntzer M, Voss M, Kroner J, Ignatius A, Fischer V. The Role of Mast Cells in Bone Metabolism and Bone Disorders. Front Immunol 2020; 11:163. [PMID: 32117297 PMCID: PMC7025484 DOI: 10.3389/fimmu.2020.00163] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 01/21/2020] [Indexed: 12/15/2022] Open
Abstract
Mast cells (MCs) are important sensor and effector cells of the immune system that are involved in many physiological and pathological conditions. Increasing evidence suggests that they also play an important role in bone metabolism and bone disorders. MCs are located in the bone marrow and secrete a wide spectrum of mediators, which can be rapidly released upon activation of mature MCs following their differentiation in mucosal or connective tissues. Many of these mediators can exert osteocatabolic effects by promoting osteoclast formation [e.g., histamine, tumor necrosis factor (TNF), interleukin-6 (IL-6)] and/or by inhibiting osteoblast activity (e.g., IL-1, TNF). By contrast, MCs could potentially act in an osteoprotective manner by stimulating osteoblasts (e.g., transforming growth factor-β) or reducing osteoclastogenesis (e.g., IL-12, interferon-γ). Experimental studies investigating MC functions in physiological bone turnover using MC-deficient mouse lines give contradictory results, reporting delayed or increased bone turnover or no influence depending on the mouse model used. By contrast, the involvement of MCs in various pathological conditions affecting bone is evident. MCs may contribute to the pathogenesis of primary and secondary osteoporosis as well as inflammatory disorders, including rheumatoid arthritis and osteoarthritis, because increased numbers of MCs were found in patients suffering from these diseases. The clinical observations could be largely confirmed in experimental studies using MC-deficient mouse models, which also provide mechanistic insights. MCs also regulate bone healing after fracture by influencing the inflammatory response toward the fracture, vascularization, bone formation, and callus remodeling by osteoclasts. This review summarizes the current view and understanding of the role of MCs on bone in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Deniz Ragipoglu
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anne Dudeck
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Melanie Haffner-Luntzer
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Martin Voss
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Jochen Kroner
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anita Ignatius
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Verena Fischer
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
20
|
Abstract
Cytokines and hematopoietic growth factors have traditionally been thought of as regulators of the development and function of immune and blood cells. However, an ever-expanding number of these factors have been discovered to have major effects on bone cells and the development of the skeleton in health and disease (Table 1). In addition, several cytokines have been directly linked to the development of osteoporosis in both animal models and in patients. In order to understand the mechanisms regulating bone cells and how this may be dysregulated in disease states, it is necessary to appreciate the diverse effects that cytokines and inflammation have on osteoblasts, osteoclasts, and bone mass. This chapter provides a broad overview of this topic with extensive references so that, if desired, readers can access specific references to delve into individual topics in greater detail.
Collapse
Affiliation(s)
- Joseph Lorenzo
- Departments of Medicine and Orthopaedic Surgery, UConn Health, Farmington, CT, USA.
| |
Collapse
|
21
|
Yongyun C, Jingwei Z, Zhiqing L, Wenxiang C, Huiwu L. Andrographolide stimulates osteoblastogenesis and bone formation by inhibiting nuclear factor kappa-Β signaling both in vivo and in vitro. J Orthop Translat 2019; 19:47-57. [PMID: 31844613 PMCID: PMC6896731 DOI: 10.1016/j.jot.2019.02.001] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/23/2019] [Accepted: 02/07/2019] [Indexed: 11/23/2022] Open
Abstract
Osteoporosis is a bone disease that is associated with a decrease in bone mineral density, deterioration of bone microarchitecture and increased fracture risk. Currently, available treatments mainly focus on either inhibiting osteoclast function, such as administration of bisphosphonate, calcitonin, oestrogen, selective oestrogen receptor modulator and so on, or stimulating osteoblasts, such as parathyroid hormone, to improve bone mass and skeletal microarchitecture. However, there is no option that is completely satisfactory because of the limitations of monotherapy with either class. Thus, it is highly appealing to investigate novel drugs with both antiresorptive and osteoanabolic activities that have the potential to be more beneficial than monotherapy because of the different mechanism of action. As has been proven in previous study that andrographolide (AP), as a key herbal medicine, could suppress osteoclast formation and function both in vivo and in vitro. The purpose of this present study was to identify the effect of AP on osteoblast differentiation and oestrogen deficiency-induced osteoporosis. It was concluded that AP significantly reduced oestrogen deficiency-induced bone loss in vivo. Furthermore, it was proved that tumor necrosis factor alpha severely impaired bone morphogenetic protein-2 (BMP-2)-induced osteoblast differentiation, and this inhibition could be greatly attenuated by AP. This was further supported by the fact that AP significantly increases the expression of osteoblast-specific markers, including runt-related transcription factor-2, osteocalcin and osteopontin. In addition, molecular analysis revealed that AP greatly ceased tumor necrosis factor alpha-mediated stimulation of nuclear factor kappa-Β activity, whereas overexpression of the nuclear factor kappa-Β subunit p65 reversed the stimulatory effects of AP on osteoblast differentiation. Thus, combined with previous study, AP was demonstrated to be a novel agent with both antiresorptive and osteoanabolic activities and had the potential to be developed as an antiosteoporosis alternative. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE This study provides strong evidence for the identification that AP has both antiresorptive and osteoanabolic activities and thus has great potential to be developed as a novel antiosteoporosis agent.
Collapse
Affiliation(s)
| | | | | | | | - Li Huiwu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People’s Republic of China
| |
Collapse
|
22
|
Soda N, Sakai N, Kato H, Takami M, Fujita T. Singleton-Merten Syndrome-like Skeletal Abnormalities in Mice with Constitutively Activated MDA5. THE JOURNAL OF IMMUNOLOGY 2019; 203:1356-1368. [PMID: 31366715 DOI: 10.4049/jimmunol.1900354] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/01/2019] [Indexed: 12/14/2022]
Abstract
Singleton-Merten syndrome (SMS) is a type I interferonopathy characterized by dental dysplasia, aortic calcification, skeletal abnormalities, glaucoma, and psoriasis. A missense mutation in IFIH1 encoding a cytoplasmic viral RNA sensor MDA5 has recently been identified in the SMS patients as well as in patients with a monogenic form of lupus. We previously reported that Ifih1gs/+ mice express a constitutively active MDA5 and spontaneously develop lupus-like nephritis. In this study, we demonstrate that the Ifih1gs/+ mice also exhibit SMS-like bone abnormalities, including decreased bone mineral density and thin cortical bone. Histological analysis revealed a low number of osteoclasts, low bone formation rate, and abnormal development of growth plate cartilages in Ifih1gs/+ mice. These abnormalities were not observed in Ifih1gs/+ ・Mavs-/- and Ifih1gs/+ ・Ifnar1-/- mice, indicating the critical role of type I IFNs induced by MDA5/MAVS-dependent signaling in the bone pathogenesis of Ifih1gs/+ mice, affecting bone turnover. Taken together, our findings suggest the inhibition of type I IFN signaling as a possible effective therapeutic strategy for bone disorders in SMS patients.
Collapse
Affiliation(s)
- Nobumasa Soda
- Laboratory of Molecular Genetics, Institute for Frontier Life and Medical Science, Kyoto University, Kyoto, 606-8507 Japan.,Laboratory of Molecular and Cellular Immunology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501 Japan
| | - Nobuhiro Sakai
- Department of Pharmacology, School of Dentistry, Showa University, Tokyo, 142-8555 Japan; and
| | - Hiroki Kato
- Laboratory of Molecular Genetics, Institute for Frontier Life and Medical Science, Kyoto University, Kyoto, 606-8507 Japan.,Institute of Cardiovascular Immunology, University Hospital Bonn, University of Bonn, Bonn, 53127 Germany
| | - Masamichi Takami
- Department of Pharmacology, School of Dentistry, Showa University, Tokyo, 142-8555 Japan; and
| | - Takashi Fujita
- Laboratory of Molecular Genetics, Institute for Frontier Life and Medical Science, Kyoto University, Kyoto, 606-8507 Japan; .,Laboratory of Molecular and Cellular Immunology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501 Japan
| |
Collapse
|
23
|
Evaluation of the Effects of Low-Level Laser Therapy on Diabetic Bone Healing. J Craniofac Surg 2019; 30:1994-1998. [PMID: 31232987 DOI: 10.1097/scs.0000000000005654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The aim of the present study was to evaluate the effects of low-level laser therapy (LLLT) and biphasic alloplastic bone graft material on diabetic bone healing. Induction of diabetes was performed in 14 male Sprague-Dawley rats by intraperitoneal injection of a 50 mg/kg dose of streptozotocin. Two bilaterally symmetrical non-critical-sized bone defects were created in the parietal bones in each rat. Right defects were filled with biphasic alloplastic bone graft. Rats were randomly divided into 2 groups, with 1 group receiving 10 sessions of LLLT (GaAlAs, 78.5 J/cm, 100mW, 0.028 cm beam). The LLLT was started immediately after surgery and once every 3 days during postoperative period. At the end of treatment period, new bone formation and osteoblast density were determined using histomorphometry. Empty (control), graft-filled, LLLT-treated and both graft-filled and LLLT-treated bone defects were compared. New bone formation was higher in the graft treatment samples compared with the control (P = 0.009) and laser samples (P = 0.029). In addition, graft-laser combination treatment samples revealed higher bone formation than control (P = 0.008) and laser (P = 0.026) samples. Osteoblast density was significantly higher in the laser treatment (P <0.001), graft treatment (P = 0.001) and graft-laser combination treatment (P <0.001) samples than control samples. In addition, significantly higher osteoblast density was observed in the graft-laser combination treatment samples compared to the graft treatment samples (P = 0.005). The LLLT was effective to stimulate osteoblastogenesis but failed to increase bone formation. Graft augmentation for treatment of bone defects seems essential for proper bone healing in diabetes, regeneration may be supported by the LLLT to enhance osteoblastogenesis.
Collapse
|
24
|
Ni C, Zhou J, Kong N, Bian T, Zhang Y, Huang X, Xiao Y, Yang W, Yan F. Gold nanoparticles modulate the crosstalk between macrophages and periodontal ligament cells for periodontitis treatment. Biomaterials 2019; 206:115-132. [DOI: 10.1016/j.biomaterials.2019.03.039] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/23/2019] [Indexed: 12/12/2022]
|
25
|
Fawzy El-Sayed KM, Elahmady M, Adawi Z, Aboushadi N, Elnaggar A, Eid M, Hamdy N, Sanaa D, Dörfer CE. The periodontal stem/progenitor cell inflammatory-regenerative cross talk: A new perspective. J Periodontal Res 2019; 54:81-94. [PMID: 30295324 DOI: 10.1111/jre.12616] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 07/24/2018] [Accepted: 08/31/2018] [Indexed: 12/13/2022]
Abstract
Adult multipotent stem/progenitor cells, with remarkable regenerative potential, have been isolated from various components of the human periodontium. These multipotent stem/progenitor cells include the periodontal ligament stem/progenitor cells (PDLSCs), stem cells from the apical papilla (SCAP), the gingival mesenchymal stem/progenitor cells (G-MSCs), and the alveolar bone proper stem/progenitor cells (AB-MSCs). Whereas inflammation is regarded as the reason for tissue damage, it also remains a fundamental step of any early healing process. In performing their periodontal tissue regenerative/reparative activity, periodontal stem/progenitor cells interact with their surrounding inflammatory micro-environmental, through their expressed receptors, which could influence their fate and the outcome of any periodontal stem/progenitor cell-mediated reparative/regenerative activity. The present review discusses the current understanding about the interaction of periodontal stem/progenitor cells with their surrounding inflammatory micro-environment, elaborates on the inflammatory factors influencing their stemness, proliferation, migration/homing, differentiation, and immunomodulatory attributes, the possible underlying intracellular mechanisms, as well as their proposed relationship to the canonical and noncanonical Wnt pathways.
Collapse
Affiliation(s)
- Karim M Fawzy El-Sayed
- Oral Medicine and Periodontology Department, Faculty of Oral and Dental Medicine, Cairo University, Cairo, Egypt
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| | | | - Zeina Adawi
- Faculty of Dentistry, New Giza University, Giza, Egypt
| | | | - Ali Elnaggar
- Faculty of Dentistry, New Giza University, Giza, Egypt
| | - Maryam Eid
- Faculty of Dentistry, New Giza University, Giza, Egypt
| | - Nayera Hamdy
- Faculty of Dentistry, New Giza University, Giza, Egypt
| | - Dalia Sanaa
- Faculty of Dentistry, New Giza University, Giza, Egypt
| | - Christof E Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| |
Collapse
|
26
|
Nazrun Shuid A, Das S, Mohamed IN. Therapeutic effect of Vitamin E in preventing bone loss: An evidence-based review. INT J VITAM NUTR RES 2019; 89:357-370. [PMID: 30856080 DOI: 10.1024/0300-9831/a000566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The present review explored the anti-inflammatory and immunomodulatory properties of vitamin E, which has protective action against osteoporosis. A systematic review of the literature was conducted to identify the published bone studies on vitamin E. The studies included inflammatory or immunology-related parameters. Medline and Scopus databases were searched for relevant studies published from 2005 till 2015. Research articles published in English and confined to the effect of vitamin E on bone were included. It is pertinent to mention that these studies took into consideration inflammatory or immunology parameters including interleukin (IL)-1, IL-6, receptor activator of nuclear factor kappa-B ligand (RANKL), inducible nitric oxide synthases (iNOS), serum amyloid A (SAA), e-selection and high-sensitivity C-reactive protein (hs-CRP). An extended literature search yielded 127 potentially relevant articles with seven articles meeting the inclusion and exclusion criteria. Another recent article was added with the total number accounting to eight. All these included literature comprised five animal studies, one in-vitro study and two human studies. These studies demonstrated that vitamin E, especially tocotrienol, was able to alleviate IL-1, IL-6, RANKL, iNOS and hs-CRP levels in relation to bone metabolism. In conclusion, vitamin E exerts its anti-osteoporotic actions via its anti-inflammatory and immunomodulatory effects.
Collapse
Affiliation(s)
- Ahmad Nazrun Shuid
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre
| | - Srijit Das
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre
| | - Isa Naina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre
| |
Collapse
|
27
|
Sun W, Meednu N, Rosenberg A, Rangel-Moreno J, Wang V, Glanzman J, Owen T, Zhou X, Zhang H, Boyce BF, Anolik JH, Xing L. B cells inhibit bone formation in rheumatoid arthritis by suppressing osteoblast differentiation. Nat Commun 2018; 9:5127. [PMID: 30510188 PMCID: PMC6277442 DOI: 10.1038/s41467-018-07626-8] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 11/06/2018] [Indexed: 02/05/2023] Open
Abstract
The function of B cells in osteoblast (OB) dysfunction in rheumatoid arthritis (RA) has not been well-studied. Here we show that B cells are enriched in the subchondral and endosteal bone marrow (BM) areas adjacent to osteocalcin+ OBs in two murine RA models: collagen-induced arthritis and the TNF-transgenic mice. Subchondral BM B cells in RA mice express high levels of OB inhibitors, CCL3 and TNF, and inhibit OB differentiation by activating ERK and NF-κB signaling pathways. The inhibitory effect of RA B cells on OB differentiation is blocked by CCL3 and TNF neutralization, and deletion of CCL3 and TNF in RA B cells completely rescues OB function in vivo, while B cell depletion attenuates bone erosion and OB inhibition in RA mice. Lastly, B cells from RA patients express CCL3 and TNF and inhibit OB differentiation, with these effects ameliorated by CCL3 and TNF neutralization. Thus, B cells inhibit bone formation in RA by producing multiple OB inhibitors.
Collapse
Affiliation(s)
- Wen Sun
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 210029, Nanjing, China
| | - Nida Meednu
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Alexander Rosenberg
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Victor Wang
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Jason Glanzman
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Teresa Owen
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Xichao Zhou
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Hengwei Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Jennifer H Anolik
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
28
|
Picke AK, Campbell GM, Schmidt FN, Busse B, Rauner M, Simon JC, Anderegg U, Hofbauer LC, Saalbach A. Thy-1 Deficiency Augments Bone Loss in Obesity by Affecting Bone Formation and Resorption. Front Cell Dev Biol 2018; 6:127. [PMID: 30333974 PMCID: PMC6176687 DOI: 10.3389/fcell.2018.00127] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/13/2018] [Indexed: 12/30/2022] Open
Abstract
Healthy bone remodeling results from a balanced bone formation and bone resorption realized by bone-forming osteoblasts and bone-resorbing osteoclasts, respectively. Recently, Thy-1 (CD90) was identified as positive regulator of osteoblast differentiation and activation, thus, promoting bone formation while concurrently inhibiting adipogenesis and obesity in mice. Additionally, Thy-1 did not affect bone resorption. An obesity-related co-morbidity that is increasing in prevalence is a disturbed bone formation resulting in an increased fracture risk. The underlying mechanisms of obesity-induced bone alterations are not yet fully elucidated and therefore therapy options for efficient bone-anabolic treatments are limited. Therefore, we investigated the impact of Thy-1 on bone metabolism under obese conditions. Indeed, high fat diet (HFD) induced obese mice lacking Thy-1 (Thy-1−/−) showed increased body fat mass compared to wildtype (WT) mice while bone mass (−38%) and formation (−57%) were decreased as shown by micro-computed tomography (μCT) measurement, histological analysis, and fourier-transform infrared spectroscopy (FTIR). Interestingly, under obese conditions, lack of Thy-1 affected both osteoblast and osteoclast function. Number (−30%) and activity of osteoblasts were decreased in obese Thy-1−/− mice while osteoclast number (+39%) and activity were increased. Facilitated bone marrow fat accumulation (+56%) in obese Thy-1−/− mice compared to obese WT mice was associated with upregulated tumor necrosis factor α (Tnfα, +46%) and colony stimulating factor 1 receptor (Csf1r) expression, strong promoters of osteoclast differentiation. Moreover, lack of Thy-1 was accompanied by a reduction of osteoprotegerin (Tnfrsf11b) expression (−36%), an inhibitor of osteoclast differentiation. Altered Tnfα, Csf1r, and Tnfrsf11b expression might be responsible for elevated osteoclast activity in obese Thy-1-deficient mice. In summary, our findings show that lack of Thy-1 promotes obesity under HFD conditions while concurrently decreasing bone mass and formation. Mechanistic studies revealed that under obese conditions lack of Thy-1 impairs both bone formation and bone resorption.
Collapse
Affiliation(s)
- Ann-Kristin Picke
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.,Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Graeme M Campbell
- Institute of Biomechanics, TUHH Hamburg University of Technology, Hamburg, Germany
| | - Felix N Schmidt
- Department of Osteology and Biomechanics, University Medical Center, Hamburg, Germany
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center, Hamburg, Germany
| | - Martina Rauner
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Jan C Simon
- Department of Dermatology, Venerology, and Allergology, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Ulf Anderegg
- Department of Dermatology, Venerology, and Allergology, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Lorenz C Hofbauer
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Anja Saalbach
- Department of Dermatology, Venerology, and Allergology, Medical Faculty, Leipzig University, Leipzig, Germany
| |
Collapse
|
29
|
Lata M, Hettinghouse AS, Liu CJ. Targeting tumor necrosis factor receptors in ankylosing spondylitis. Ann N Y Acad Sci 2018; 1442:5-16. [PMID: 30008173 DOI: 10.1111/nyas.13933] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/19/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023]
Abstract
Over the past two decades, considerable advances in our understanding of inflammatory and immune pathways have allowed for the growing use of targeted biologic therapy. Most notably, the introduction of tumor necrosis factor (TNF) inhibitors has dramatically changed the management of autoimmune inflammatory disorders, including ankylosing spondylitis (AS). Despite the efficacy of TNF inhibitors documented in multiple clinical trials, anti-TNF therapy in AS is far from foolproof; it is associated with serious adverse effects and limited response to therapy in some patients. Moreover, specific questions regarding the role of TNF as a mediator of AS remain unanswered. Therefore, additional efforts are needed in order to better understand the role of TNF in the pathogenesis of AS and to develop safer and more effective treatment strategies. The purpose of this review is to better the understanding of the physiologic and pathogenic roles of TNF signaling in the course of AS. Relevant TNF biology and novel approaches to TNF blockade in AS are discussed.
Collapse
Affiliation(s)
- Michal Lata
- Department of Orthopedic Surgery, New York University Medical Center, Hospital for Joint Diseases, New York, New York
| | - Aubryanna S Hettinghouse
- Department of Orthopedic Surgery, New York University Medical Center, Hospital for Joint Diseases, New York, New York
| | - Chuan-Ju Liu
- Department of Orthopedic Surgery, New York University Medical Center, Hospital for Joint Diseases, New York, New York.,Department of Cell Biology, New York University School of Medicine, New York, New York
| |
Collapse
|
30
|
Araya HF, Sepulveda H, Lizama CO, Vega OA, Jerez S, Briceño PF, Thaler R, Riester SM, Antonelli M, Salazar-Onfray F, Rodríguez JP, Moreno RD, Montecino M, Charbonneau M, Dubois CM, Stein GS, van Wijnen AJ, Galindo MA. Expression of the ectodomain-releasing protease ADAM17 is directly regulated by the osteosarcoma and bone-related transcription factor RUNX2. J Cell Biochem 2018; 119:8204-8219. [PMID: 29923217 DOI: 10.1002/jcb.26832] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 03/06/2018] [Indexed: 01/04/2023]
Abstract
Osteoblast differentiation is controlled by transcription factor RUNX2 which temporally activates or represses several bone-related genes, including those encoding extracellular matrix proteins or factors that control cell-cell, and cell-matrix interactions. Cell-cell communication in the many skeletal pericellular micro-niches is critical for bone development and involves paracrine secretion of growth factors and morphogens. This paracrine signaling is in part regulated by "A Disintegrin And Metalloproteinase" (ADAM) proteins. These cell membrane-associated metalloproteinases support proteolytic release ("shedding") of protein ectodomains residing at the cell surface. We analyzed microarray and RNA-sequencing data for Adam genes and show that Adam17, Adam10, and Adam9 are stimulated during BMP2 mediated induction of osteogenic differentiation and are robustly expressed in human osteoblastic cells. ADAM17, which was initially identified as a tumor necrosis factor alpha (TNFα) converting enzyme also called (TACE), regulates TNFα-signaling pathway, which inhibits osteoblast differentiation. We demonstrate that Adam17 expression is suppressed by RUNX2 during osteoblast differentiation through the proximal Adam17 promoter region (-0.4 kb) containing two functional RUNX2 binding motifs. Adam17 downregulation during osteoblast differentiation is paralleled by increased RUNX2 expression, cytoplasmic-nuclear translocation and enhanced binding to the Adam17 proximal promoter. Forced expression of Adam17 reduces Runx2 and Alpl expression, indicating that Adam17 may negatively modulate osteoblast differentiation. These findings suggest a novel regulatory mechanism involving a reciprocal Runx2-Adam17 negative feedback loop to regulate progression through osteoblast differentiation. Our results suggest that RUNX2 may control paracrine signaling through regulation of ectodomain shedding at the cell surface of osteoblasts by directly suppressing Adam17 expression.
Collapse
Affiliation(s)
- Héctor F Araya
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Hugo Sepulveda
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Carlos O Lizama
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar A Vega
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Sofia Jerez
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pedro F Briceño
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Scott M Riester
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Marcelo Antonelli
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile.,Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan Pablo Rodríguez
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), University of Chile, Santiago, Chile
| | - Ricardo D Moreno
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Martin Montecino
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Martine Charbonneau
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Claire M Dubois
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Gary S Stein
- Department of Biochemistry and University of Vermont Cancer Center, The Robert Larner MD College of Medicine, University of Vermont, Burlington, Vermont
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Mario A Galindo
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
31
|
Paine A, Ritchlin C. Altered Bone Remodeling in Psoriatic Disease: New Insights and Future Directions. Calcif Tissue Int 2018; 102:559-574. [PMID: 29330560 PMCID: PMC5906143 DOI: 10.1007/s00223-017-0380-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/27/2017] [Indexed: 12/15/2022]
Abstract
Psoriatic arthritis (PsA) is an inflammatory rheumatic disorder that occurs in patients with psoriasis and predominantly affects musculoskeletal structures, skin, and nails. The etiology of PsA is not well understood but evidence supports an interplay of genetic, immunologic, and environmental factors which promote pathological bone remodeling and joint damage in PsA. Localized and systemic bone loss due to increased activity of osteoclasts is well established in PsA based on animal models and translational studies. In contrast, the mechanisms responsible for pathological bone remodeling in PsA remain enigmatic although new candidate molecules and pathways have been identified. Recent reports have revealed novel findings related to bone erosion and pathologic bone formation in PsA. Many associated risk factors and contributing molecular mechanisms have also been identified. In this review, we discuss new developments in the field, point out unresolved questions regarding the pathogenetic origins of the wide array of bone phenotypes in PsA, and discuss new directions for investigation.
Collapse
Affiliation(s)
- Ananta Paine
- Allergy, Immunology & Rheumatology Division, University of Rochester Medical Center, Rochester, NY, 14623, USA.
| | - Christopher Ritchlin
- Allergy, Immunology & Rheumatology Division, University of Rochester Medical Center, Rochester, NY, 14623, USA
| |
Collapse
|
32
|
Zhu S, He H, Gao C, Luo G, Xie Y, Wang H, Tian L, Chen X, Yu X, He C. Ovariectomy-induced bone loss in TNFα and IL6 gene knockout mice is regulated by different mechanisms. J Mol Endocrinol 2018; 60:185-198. [PMID: 29339399 DOI: 10.1530/jme-17-0218] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 01/15/2018] [Indexed: 02/05/2023]
Abstract
We examined the effects of tumor necrosis factor-α (TNFα) and interleukin-6 (IL6) gene knockout in preserving the bone loss induced by ovariectomy (OVX) and the mechanisms involved in bone metabolism. Twenty female wild-type (WT), TNFα-knockout (TNFα-/-) or IL6-knockout (IL6-/-) mice aged 12 weeks were sham-operated (SHAM) or subjected to OVX and killed after 4 weeks. Bone mass and skeletal microarchitecture were determined using micro-CT. Bone marrow stromal cells (BMSCs) from all three groups (WT, TNFα-/- and IL6-/-) were induced to differentiate into osteoblasts or osteoclasts and treated with 17-β-estradiol. Bone metabolism was assessed by histological analysis, serum analyses and qRT-PCR. OVX successfully induced a high turnover in all mice, but a repair effect was observed in TNFα-/- and IL6-/- mice. The ratio of femoral trabecular bone volume to tissue volume, trabecular number and trabecular thickness were significantly decreased in WT mice subjected to OVX, but increased in TNFα-/- mice (1.62, 1.34, 0.27-fold respectively; P < 0.01) and IL6-/- mice (1.34, 0.80, 0.22-fold respectively; P < 0.01). Furthermore, we observed a 29.6% increase in the trabecular number in TNFα-/- mice when compared to the IL6-/- mice. Both, TNFα-/- and IL6-/- BMSCs exhibited decreased numbers of TRAP-positive cells and an increase in ALP-positive cells, with or without E2 treatment (P < 0.05). While the knockout of TNFα or IL6 significantly upregulated mRNA expressions of osteoblast-related genes (Runx2 and Col1a1) and downregulated osteoclast-related mRNA for TRAP, MMP9 and CTSK in vivo and in vitro, TNFα knockout appeared to have roles beyond IL6 knockout in upregulating Col1a1 mRNA expression and downregulating mRNA expressions of WNT-related genes (DKK1 and Sost) and TNF-related activation-induced genes (TRAF6). TNFα seemed to be more potentially invasive in inhibiting bone formation and enhancing TRAF6-mediated osteoclastogenesis than IL6, implying that the regulatory mechanisms of TNFα and IL6 in bone metabolism may be different.
Collapse
Affiliation(s)
- Siyi Zhu
- Rehabilitation Medicine CenterWest China Hospital, Sichuan University, Chengdu, China
- Rehabilitation Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University, Chengdu, China
- Laboratory of Endocrinology and MetabolismDepartment of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongchen He
- Rehabilitation Medicine CenterWest China Hospital, Sichuan University, Chengdu, China
| | - Chengfei Gao
- Rehabilitation Medicine CenterWest China Hospital, Sichuan University, Chengdu, China
- Rehabilitation Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University, Chengdu, China
- Institute for Disaster Management and ReconstructionSichuan University-The Hong Kong Polytechnic University, Chengdu, China
| | - Guojing Luo
- Laboratory of Endocrinology and MetabolismDepartment of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Xie
- Laboratory of Endocrinology and MetabolismDepartment of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Haiming Wang
- Rehabilitation Medicine CenterWest China Hospital, Sichuan University, Chengdu, China
- Rehabilitation Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University, Chengdu, China
| | - Li Tian
- Laboratory of Endocrinology and MetabolismDepartment of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Chen
- Laboratory of Endocrinology and MetabolismDepartment of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijie Yu
- Laboratory of Endocrinology and MetabolismDepartment of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chengqi He
- Rehabilitation Medicine CenterWest China Hospital, Sichuan University, Chengdu, China
- Rehabilitation Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University, Chengdu, China
- Institute for Disaster Management and ReconstructionSichuan University-The Hong Kong Polytechnic University, Chengdu, China
| |
Collapse
|
33
|
Wei F, Xiao Y. Modulation of the Osteoimmune Environment in the Development of Biomaterials for Osteogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1077:69-86. [DOI: 10.1007/978-981-13-0947-2_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
34
|
Wang Y, Dai J, Zhu Y, Zhong W, Lu S, Chen H, Chai Y. Paeoniflorin regulates osteoclastogenesis and osteoblastogenesis via manipulating NF-κB signaling pathway both in vitro and in vivo. Oncotarget 2017; 9:7372-7388. [PMID: 29484117 PMCID: PMC5800909 DOI: 10.18632/oncotarget.23677] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/16/2017] [Indexed: 12/21/2022] Open
Abstract
The metabolic balance between synthesis and resorption of the bone is maintained by osteoblasts and osteoclasts, respectively. Identification of agents that stimulate bone formation and suppress excessive osteoclast formation, may aid in preventing and treating conditions like osteoporosis and periprosthetic loosening. Paeoniflorin is a natural product derived from Paeonia lactiflora Pall with anti-inflammatory, analgesic, and diuretic properties. However, the effect of paeoniflorin on osteoclastogenesis and osteoblastogenesis is unknown. Herein, we demonstrated that paeoniflorin has a dose-dependent suppressive action on RANKL-evoked osteoclast differentiation and bone resorption, achieved by inhibiting the NF-κB pathway and subunit p65 nuclear translocation. Simultaneously, paeoniflorin was also found to stimulate osteoblast differentiation and bone mineralization, in addition to rescuing TNFα-impaired osteoblastogenesis. At the molecular level, paeoniflorin was found to inhibit NF-κB transcriptional activity and stimulate osteoblastogenesis-related marker gene expression (ALP, osteocalcin, OPN and Runx2), a trend that was inhibited by p65 overexpression. In ovariectomized mice, paeoniflorin was found to improve osteoblast activity, inhibit osteoclast activity, and thus, reduce ovariectomy-induced osteoporosis. Our study demonstrated that paeoniflorin simultaneously suppressed osteoclastogenesis and facilitated osteoblastogenesis by manipulating the actions of NF-κB. Therefore, paeoniflorin may serve as an ideal therapeutic antidote for osteoporosis.
Collapse
Affiliation(s)
- Yanmao Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, PR China
| | - Jiezhi Dai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, PR China
| | - Yu Zhu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, PR China
| | - Wanrun Zhong
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, PR China
| | - Shengdi Lu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, PR China
| | - Hua Chen
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, PR China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, PR China
| |
Collapse
|
35
|
Cirillo F, Lazzeroni P, Sartori C, Street ME. Inflammatory Diseases and Growth: Effects on the GH-IGF Axis and on Growth Plate. Int J Mol Sci 2017; 18:E1878. [PMID: 28858208 PMCID: PMC5618527 DOI: 10.3390/ijms18091878] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 08/23/2017] [Accepted: 08/29/2017] [Indexed: 02/08/2023] Open
Abstract
This review briefly describes the most common chronic inflammatory diseases in childhood, such as cystic fibrosis (CF), inflammatory bowel diseases (IBDs), juvenile idiopathic arthritis (JIA), and intrauterine growth restriction (IUGR) that can be considered, as such, for the changes reported in the placenta and cord blood of these subjects. Changes in growth hormone (GH) secretion, GH resistance, and changes in the insulin-like growth factor (IGF) system are described mainly in relationship with the increase in nuclear factor-κB (NF-κB) and pro-inflammatory cytokines. Changes in the growth plate are also reported as well as a potential role for microRNAs (miRNAs) and thus epigenetic changes in chronic inflammation. Many mechanisms leading to growth failure are currently known; however, it is clear that further research in the field is still warranted.
Collapse
Affiliation(s)
- Francesca Cirillo
- Division of Paediatric Endocrinology and Diabetology, Department of Obstetrics, Gynaecology and Paediatrics, Azienda AUSL-IRCCS, Viale Risorgimento, 80, 42123 Reggio Emilia, Italy.
| | - Pietro Lazzeroni
- Division of Paediatric Endocrinology and Diabetology, Department of Obstetrics, Gynaecology and Paediatrics, Azienda AUSL-IRCCS, Viale Risorgimento, 80, 42123 Reggio Emilia, Italy.
| | - Chiara Sartori
- Division of Paediatric Endocrinology and Diabetology, Department of Obstetrics, Gynaecology and Paediatrics, Azienda AUSL-IRCCS, Viale Risorgimento, 80, 42123 Reggio Emilia, Italy.
| | - Maria Elisabeth Street
- Division of Paediatric Endocrinology and Diabetology, Department of Obstetrics, Gynaecology and Paediatrics, Azienda AUSL-IRCCS, Viale Risorgimento, 80, 42123 Reggio Emilia, Italy.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW In the process of bone fracture healing, inflammation is thought to be an essential process that precedes bone formation and remodeling. We review recent studies on bone fracture healing from an osteoimmunological point of view. RECENT FINDINGS Based on previous observations that many types of immune cells infiltrate into the bone injury site and release a variety of molecules, recent studies have addressed the roles of specific immune cell subsets. Macrophages and interleukin (IL)-17-producing γδ T cells enhance bone healing, whereas CD8+ T cells impair bone repair. Additionally, IL-10-producing B cells may contribute to bone healing by suppressing excessive and/or prolonged inflammation. Although the involvement of other cells and molecules has been suggested, the precise underlying mechanisms remain elusive. Accumulating evidence has begun to reveal the deeper picture of bone fracture healing. Further studies are required for the development of novel therapeutic strategies for bone fracture.
Collapse
Affiliation(s)
- Takehito Ono
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
37
|
Barrachina L, Remacha AR, Romero A, Vázquez FJ, Albareda J, Prades M, Ranera B, Zaragoza P, Martín-Burriel I, Rodellar C. Inflammation affects the viability and plasticity of equine mesenchymal stem cells: possible implications in intra-articular treatments. J Vet Sci 2017; 18:39-49. [PMID: 27297420 PMCID: PMC5366301 DOI: 10.4142/jvs.2017.18.1.39] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/19/2016] [Accepted: 05/12/2016] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are gaining relevance for treating equine joint injuries because of their ability to limit inflammation and stimulate regeneration. Because inflammation activates MSC immunoregulatory function, proinflammatory priming could improve MSC efficacy. However, inflammatory molecules present in synovial fluid or added to the culture medium might have deleterious effects on MSCs. Therefore, this study was conducted to investigate the effects of inflammatory synovial fluid and proinflammatory cytokines priming on viability and plasticity of equine MSCs. Equine bone marrow derived MSCs (eBM-MSCs) from three animals were cultured for 72 h in media supplemented with: 20% inflammatory synovial fluid (SF); 50 ng/mL IFN-γ and TNF-α (CK50); and 20 ng/mL IFN-γ and TNF-α (CK20). Proliferation assay and expression of proliferation and apoptosis-related genes showed that SF exposed-eBM-MSCs maintained their viability, whereas the viability of CK primed-eBM-MSCs was significantly impaired. Tri-lineage differentiation assay revealed that exposure to inflammatory synovial fluid did not alter eBM-MSCs differentiation potential; however, eBM-MSCs primed with cytokines did not display osteogenic, adipogenic or chondrogenic phenotype. The inflammatory synovial environment is well tolerated by eBM-MSCs, whereas cytokine priming negatively affects the viability and differentiation abilities of eBM-MSCs, which might limit their in vivo efficacy.
Collapse
Affiliation(s)
- Laura Barrachina
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain.,Service of Equine Surgery and Medicine, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Ana Rosa Remacha
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Antonio Romero
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain.,Service of Equine Surgery and Medicine, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Francisco José Vázquez
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain.,Service of Equine Surgery and Medicine, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Jorge Albareda
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain.,Service of Orthopedic Surgery and Traumatology, University Clinical Hospital Lozano Blesa, 50009 Zaragoza, Spain
| | - Marta Prades
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain.,Service of Equine Surgery, Veterinary Hospital, Autonomous University of Barcelona, 08193 Barcelona, Spain
| | - Beatriz Ranera
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Pilar Zaragoza
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Inmaculada Martín-Burriel
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| | - Clementina Rodellar
- Laboratory of Biochemical Genetics LAGENBIO, Veterinary Hospital, University of Zaragoza, 50013 Zaragoza, Spain
| |
Collapse
|
38
|
Su YW, Zhou XF, Foster BK, Grills BL, Xu J, Xian CJ. Roles of neurotrophins in skeletal tissue formation and healing. J Cell Physiol 2017; 233:2133-2145. [PMID: 28370021 DOI: 10.1002/jcp.25936] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 12/21/2022]
Abstract
Neurotrophins and their receptors are key molecules that are known to be critical in regulating nervous system development and maintenance and have been recognized to be also involved in regulating tissue formation and healing in skeletal tissues. Studies have shown that neurotrophins and their receptors are widely expressed in skeletal tissues, implicated in chondrogenesis, osteoblastogenesis, and osteoclastogenesis, and are also involved in regulating tissue formation and healing events in skeletal tissue. Increased mRNA expression for neurotrophins NGF, BDNF, NT-3, and NT-4, and their Trk receptors has been observed in injured bone tissues, and NT-3 and its receptor, TrkC, have been identified to have the highest induction at the injury site in a drill-hole injury repair model in both bone and the growth plate. In addition, NT-3 has also recently been shown to be both an osteogenic and angiogenic factor, and this neurotrophin can also enhance expression of the key osteogenic factor, BMP-2, as well as the major angiogenic factor, VEGF, to promote bone formation, vascularization, and healing of the injury site. Further studies, however, are needed to investigate if different neurotrophins have differential roles in skeletal repair, and if NT-3 can be a potential target of intervention for promoting bone fracture healing.
Collapse
Affiliation(s)
- Yu-Wen Su
- Sansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Xin-Fu Zhou
- Sansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Bruce K Foster
- Department of Orthopaedic Surgery, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Brian L Grills
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Cory J Xian
- Sansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
39
|
Lin TH, Pajarinen J, Lu L, Nabeshima A, Cordova LA, Yao Z, Goodman SB. NF-κB as a Therapeutic Target in Inflammatory-Associated Bone Diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 107:117-154. [PMID: 28215222 DOI: 10.1016/bs.apcsb.2016.11.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Inflammation is a defensive mechanism for pathogen clearance and maintaining tissue homeostasis. In the skeletal system, inflammation is closely associated with many bone disorders including fractures, nonunions, periprosthetic osteolysis (bone loss around orthopedic implants), and osteoporosis. Acute inflammation is a critical step for proper bone-healing and bone-remodeling processes. On the other hand, chronic inflammation with excessive proinflammatory cytokines disrupts the balance of skeletal homeostasis involving osteoblastic (bone formation) and osteoclastic (bone resorption) activities. NF-κB is a transcriptional factor that regulates the inflammatory response and bone-remodeling processes in both bone-forming and bone-resorption cells. In vitro and in vivo evidences suggest that NF-κB is an important potential therapeutic target for inflammation-associated bone disorders by modulating inflammation and bone-remodeling process simultaneously. The challenges of NF-κB-targeting therapy in bone disorders include: (1) the complexity of canonical and noncanonical NF-κB pathways; (2) the fundamental roles of NF-κB-mediated signaling for bone regeneration at earlier phases of tissue damage and acute inflammation; and (3) the potential toxic effects on nontargeted cells such as lymphocytes. Recent developments of novel inhibitors with differential approaches to modulate NF-κB activity, and the controlled release (local) or bone-targeting drug delivery (systemic) strategies, have largely increased the translational application of NF-κB therapy in bone disorders. Taken together, temporal modulation of NF-κB pathways with the combination of recent advanced bone-targeting drug delivery techniques is a highly translational strategy to reestablish homeostasis in the skeletal system.
Collapse
Affiliation(s)
- T-H Lin
- Stanford University, Stanford, CA, United States
| | - J Pajarinen
- Stanford University, Stanford, CA, United States
| | - L Lu
- Stanford University, Stanford, CA, United States
| | - A Nabeshima
- Stanford University, Stanford, CA, United States
| | - L A Cordova
- Stanford University, Stanford, CA, United States; Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Z Yao
- Stanford University, Stanford, CA, United States
| | - S B Goodman
- Stanford University, Stanford, CA, United States.
| |
Collapse
|
40
|
Inhibition of Runx2 signaling by TNF-α in ST2 murine bone marrow stromal cells undergoing osteogenic differentiation. In Vitro Cell Dev Biol Anim 2016; 52:1026-1033. [PMID: 27401008 DOI: 10.1007/s11626-016-0068-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 06/22/2016] [Indexed: 12/25/2022]
Abstract
Tumor necrosis factor-alpha (TNF-α) inhibits osteogenic differentiation of murine bone marrow stromal cells, and transcription factor Runx2 serves as an essential regulation target in the process. The underlying mechanism may involve the regulation of Runx2 expression and the Runx2 activity in downstream gene transcription, which has not been fully elucidated. In this study, ST2 murine bone marrow-derived stromal cells were treated with bone morphogenetic protein-2 (BMP-2) and/or TNF-α in osteogenic medium, and the expression of Runx2 was estimated. Cells were transfected with Runx2, p65, inhibitor of κBα (IκBα), 9.0 kb bone sialoprotein (BSP) promoter-luciferase or osteoblast-specific cis-acting element 2 (OSE2)-luciferase reporter vectors, and then real time-PCR and dual luciferase analysis were used to investigate the effect of TNF-α on Runx2-activated osteogenic gene transcription and the molecular mechanism. We found that TNF-α inhibited BMP-2-induced osteogenic marker expression and both the spontaneous and BMP-2-induced Runx2 expression. TNF-α stimulation or overexpression of nuclear factor-kappa B (NF-κB) p65 subunit repressed the Runx2-activated BSP and osteocalcin (OC) transcriptions. The Runx2-induced 9.0 kb BSP promoter activity was attenuated by TNF-α or p65, while the OSE2 activity was not affected. Besides, blockage of NF-κB by IκBα overexpression eliminated these inhibitory effects of TNF-α on Runx2 signaling. These results suggest that in murine bone marrow stromal cells undergoing osteogenic differentiation, TNF-α and it activated NF-κB pathway inhibit the expression of Runx2 gene, and suppress the Runx2-mediated osteogenic gene transcription via the 9.0 kb BSP promoter.
Collapse
|
41
|
Algate K, Haynes DR, Bartold PM, Crotti TN, Cantley MD. The effects of tumour necrosis factor-α on bone cells involved in periodontal alveolar bone loss; osteoclasts, osteoblasts and osteocytes. J Periodontal Res 2015; 51:549-66. [DOI: 10.1111/jre.12339] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2015] [Indexed: 12/22/2022]
Affiliation(s)
- K. Algate
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - D. R. Haynes
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - P. M. Bartold
- School of Dentistry; University of Adelaide; Adelaide SA Australia
| | - T. N. Crotti
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - M. D. Cantley
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
- Myeloma Research Laboratory; University of Adelaide; Adelaide SA Australia
| |
Collapse
|
42
|
Li J, Liu X, Zuo B, Zhang L. The Role of Bone Marrow Microenvironment in Governing the Balance between Osteoblastogenesis and Adipogenesis. Aging Dis 2015; 7:514-25. [PMID: 27493836 DOI: 10.14336/ad.2015.1206] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 12/06/2015] [Indexed: 01/08/2023] Open
Abstract
In the adult bone marrow, osteoblasts and adipocytes share a common precursor called mesenchymal stem cells (MSCs). The plasticity between the two lineages has been confirmed over the past decades, and has important implications in the etiology of bone diseases such as osteoporosis, which involves an imbalance between osteoblasts and adipocytes. The commitment and differentiation of bone marrow (BM) MSCs is tightly controlled by the local environment that maintains a balance between osteoblast lineage and adipocyte. However, pathological conditions linked to osteoporosis can change the BM microenvironment and shift the MSC fate to favor adipocytes over osteoblasts, and consequently decrease bone mass with marrow fat accumulation. This review discusses the changes that occur in the BM microenvironment under pathological conditions, and how these changes affect MSC fate. We suggest that manipulating local environments could have therapeutic implications to avoid bone loss in diseases like osteoporosis.
Collapse
Affiliation(s)
- Jiao Li
- 1Department of Cell Biology, Zunyi Medical College, Zunyi, China
| | - Xingyu Liu
- 1Department of Cell Biology, Zunyi Medical College, Zunyi, China
| | - Bin Zuo
- 2Department of Orthopedic Surgery, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Li Zhang
- 3Department of Orthopedics, Tenth People's Hospital, Shanghai Tong Ji University, School of Medicine, Shanghai, China
| |
Collapse
|
43
|
The Effect of Tumor Necrosis Factor-α at Different Concentrations on Osteogenetic Differentiation of Bone Marrow Mesenchymal Stem Cells. J Craniofac Surg 2015; 26:2081-5. [DOI: 10.1097/scs.0000000000001971] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
44
|
Diabetes mellitus related bone metabolism and periodontal disease. Int J Oral Sci 2015; 7:63-72. [PMID: 25857702 PMCID: PMC4817554 DOI: 10.1038/ijos.2015.2] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2014] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus and periodontal disease are chronic diseases affecting a large number of populations worldwide. Changed bone metabolism is one of the important long-term complications associated with diabetes mellitus. Alveolar bone loss is one of the main outcomes of periodontitis, and diabetes is among the primary risk factors for periodontal disease. In this review, we summarise the adverse effects of diabetes on the periodontium in periodontitis subjects, focusing on alveolar bone loss. Bone remodelling begins with osteoclasts resorbing bone, followed by new bone formation by osteoblasts in the resorption lacunae. Therefore, we discuss the potential mechanism of diabetes-enhanced bone loss in relation to osteoblasts and osteoclasts.
Collapse
|
45
|
Yang Y, Dai M. Expression of PADI4 in patients with ankylosing spondylitis and its role in mediating the effects of TNF-α on the proliferation and osteogenic differentiation of human mesenchymal stem cells. Int J Mol Med 2015; 36:565-70. [PMID: 26082376 DOI: 10.3892/ijmm.2015.2248] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/09/2015] [Indexed: 01/18/2023] Open
Abstract
Peptidyl arginine deiminase, type IV (PADI4) plays an important role in inflammation and in the immune response, and it has been shown to be associated with rheumatoid arthritis, osteoarthritis and ankylosing spondylitis (AS). However, little is known about the precise role of PADI4 in the pathogenic process in vitro. In this study, we aimed to investigate the expression of PADI4 in the synovial tissue of patients with AS and to determine the potential effects of PADI4 on human mesenchymal stem cell (hMSC) proliferation and osteogenic differentiation under normal and pathological conditions. Synovial tissues were collected from 18 patients with AS and 11 control subjects. The results of reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis revealed that the expression of PADI4 was upregulated in the patients with AS. In the hMSCs, the protein expression of PADI4 was increased following treatment with tumor necrosis factor-α (TNF-α) in a dose- and time-dependent manner. MTT assay revealed that TNF-α promoted hMSC proliferation. In addition, we found that TNF-α promoted the osteogenic differentiation of hMSCs, as demonstrated by an increase in alkaline phosphatase (ALP) activity, as well as an increase in the expression of bone morphogenetic protein 2 (BMP-2), runt-related transcription factor 2 (Runx2) and Osterix. The hMSCs were transfected with PADI4 siRNA to silence PADI4 expression. We found that, under normal conditions, the silencing of PADI4 did not have any effect on hMSC proliferation or osteogenic differentiation. However, in the presence of TNF-α, hMSC proliferation and osteogenic differentiation were induced. These effects were attenuated by the silencing of PADI4. In conclusion, the findings of this study demonstrate that the expression of PADI4 differs between patients with AS and normal subjects. In addition, our data suggest that PADI4 plays a role in hMSC proliferation and differentiation, which are induced by TNF-α.
Collapse
Affiliation(s)
- Yong Yang
- Department of Orthopaedics, Xinyu Hospital of Nanchang University, Xinyu, Jiangxi 338025, P.R. China
| | - Min Dai
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Nangchang, Jiangxi 330006, P.R. China
| |
Collapse
|
46
|
Impact of ellagic acid in bone formation after tooth extraction: an experimental study on diabetic rats. ScientificWorldJournal 2014; 2014:908098. [PMID: 25485304 PMCID: PMC4251085 DOI: 10.1155/2014/908098] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 08/05/2014] [Accepted: 08/26/2014] [Indexed: 01/10/2023] Open
Abstract
Objectives. To estimate the impact of ellagic acid (EA) towards healing tooth socket in diabetic animals, after tooth extraction. Methods. Twenty-four Sprague Dawley male rats weighing 250–300 g were selected for this study. All animals were intraperitoneally injected with 45 mg/kg (b.w.) of freshly prepared streptozotocin (STZ), to induce diabetic mellitus. Then, the animals were anesthetized, and the upper left central incisor was extracted and the whole extracted sockets were filled with Rosuvastatin (RSV). The rats were separated into three groups, comprising 8 rats each. The first group was considered as normal control group and orally treated with normal saline. The second group was regarded as diabetic control group and orally treated with normal saline, whereas the third group comprised diabetic rats, administrated with EA (50 mg/kg) orally. The maxilla tissue stained by eosin and hematoxylin (H&E) was used for histological examinations and immunohistochemical technique. Fibroblast growth factor (FGF-2) and alkaline phosphatase (ALP) were used to evaluate the healing process in the extracted tooth socket by immunohistochemistry test. Results. The reactions of immunohistochemistry for FGF-2 and ALP presented stronger expression, predominantly in EA treated diabetic rat, than the untreated diabetic rat. Conclusion. These findings suggest that the administration of EA combined with RSV may have accelerated the healing process of the tooth socket of diabetic rats, after tooth extraction.
Collapse
|
47
|
Cytokine-mediated bone destruction in rheumatoid arthritis. J Immunol Res 2014; 2014:263625. [PMID: 25295284 PMCID: PMC4176903 DOI: 10.1155/2014/263625] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/27/2014] [Indexed: 12/29/2022] Open
Abstract
Bone homeostasis, which involves formation and resorption, is an important process for maintaining adequate bone mass in humans. Rheumatoid arthritis (RA) is an autoimmune disease characterized by inflammation and bone loss, leading to joint destruction and deformity, and is a representative disease of disrupted bone homeostasis. The bone loss and joint destruction are mediated by immunological insults by proinflammatory cytokines and various immune cells. The connection between bone and immunity has been intensely studied and comprises the emerging field of osteoimmunology. Osteoimmunology is an interdisciplinary science investigating the interplay between the skeletal and the immune systems. The main contributors in osteoimmunology are the bone effector cells, such as osteoclasts or osteoblasts, and the immune cells, particularly lymphocytes and monocytes. Physiologically, osteoclasts originate from immune cells, and immune cells regulate osteoblasts and vice versa. Pathological conditions such as RA might affect these interactions, thereby altering bone homeostasis, resulting in the unfavorable outcome of bone destruction. In this review, we describe the osteoclastogenic roles of the proinflammatory cytokines and immune cells that are important in the pathophysiology of RA.
Collapse
|
48
|
Salamon A, Adam S, Rychly J, Peters K. Long-term tumor necrosis factor treatment induces NFκB activation and proliferation, but not osteoblastic differentiation of adipose tissue-derived mesenchymal stem cells in vitro. Int J Biochem Cell Biol 2014; 54:149-62. [PMID: 25066315 DOI: 10.1016/j.biocel.2014.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 07/16/2014] [Accepted: 07/17/2014] [Indexed: 01/08/2023]
Abstract
The pro-inflammatory cytokine tumor necrosis factor (TNF) is well known to induce differentiation of bone matrix-resorbing osteoclasts from hematopoietic stem cells. However, the impact of TNF on differentiation of bone matrix-forming osteoblasts from mesenchymal stem cells (MSC) was only fragmentarily studied so far. Therefore, we investigated what impact long-term TNF treatment has on osteoblastic differentiation of MSC isolated from the adipose tissue (ASC) in vitro. In summary, we found continuous TNF exposure to induce the nuclear factor of kappa B pathway in ASC as well as secretion of the pro-inflammatory chemokine interleukin 8, but not the mitogen-activated protein kinase and the apoptosis pathway in ASC. Moreover, TNF neither induced nor inhibited osteoblastic differentiation of ASC, but strongly increased their proliferation rate. In that manner, pro-inflammatory conditions in vivo may generate significantly increased numbers of progenitor cells, and ASC especially, in conjunction with external stimuli, may contribute to the events of ectopic ossification observed in chronic inflammatory diseases. The substantiation of the translation of our in vitro findings to the disease context encourages further in vivo studies.
Collapse
Affiliation(s)
- Achim Salamon
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany.
| | - Stefanie Adam
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany
| | - Joachim Rychly
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany
| | - Kirsten Peters
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, D-18057 Rostock, Germany
| |
Collapse
|
49
|
Yao Z, Li Y, Yin X, Dong Y, Xing L, Boyce BF. NF-κB RelB negatively regulates osteoblast differentiation and bone formation. J Bone Miner Res 2014; 29:866-77. [PMID: 24115294 PMCID: PMC3961566 DOI: 10.1002/jbmr.2108] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 09/08/2013] [Accepted: 09/19/2013] [Indexed: 11/08/2022]
Abstract
RelA-mediated NF-κB canonical signaling promotes mesenchymal progenitor cell (MPC) proliferation, but inhibits differentiation of mature osteoblasts (OBs) and thus negatively regulates bone formation. Previous studies suggest that NF-κB RelB may also negatively regulate bone formation through noncanonical signaling, but they involved a complex knockout mouse model, and the molecular mechanisms involved were not investigated. Here, we report that RelB(-/-) mice develop age-related increased trabecular bone mass associated with increased bone formation. RelB(-/-) bone marrow stromal cells expanded faster in vitro and have enhanced OB differentiation associated with increased expression of the osteoblastogenic transcription factor, Runt-related transcription factor 2 (Runx2). In addition, RelB directly targeted the Runx2 promoter to inhibit its activation. Importantly, RelB(-/-) bone-derived MPCs formed bone more rapidly than wild-type cells after they were injected into a murine tibial bone defect model. Our findings indicate that RelB negatively regulates bone mass as mice age and limits bone formation in healing bone defects, suggesting that inhibition of RelB could reduce age-related bone loss and enhance bone repair.
Collapse
Affiliation(s)
- Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yanyun Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Xiaoxiang Yin
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yufeng Dong
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Brendan F. Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
50
|
Deregulation of bone forming cells in bone diseases and anabolic effects of strontium-containing agents and biomaterials. BIOMED RESEARCH INTERNATIONAL 2014; 2014:814057. [PMID: 24800251 PMCID: PMC3988913 DOI: 10.1155/2014/814057] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 02/20/2014] [Accepted: 03/03/2014] [Indexed: 11/30/2022]
Abstract
Age-related bone loss and osteoporosis are associated with bone remodeling changes that are featured with decreased trabecular and periosteal bone formation relative to bone resorption. Current anticatabolic therapies focusing on the inhibition of bone resorption may not be sufficient in the prevention or reversal of age-related bone deterioration and there is a big need in promoting osteoblastogenesis and bone formation. Enhanced understanding of the network formed by key signaling pathways and molecules regulating bone forming cells in health and diseases has therefore become highly significant. The successful development of agonist/antagonist of the PTH and Wnt signaling pathways are profits of the understanding of these key pathways. As the core component of an approved antiosteoporosis agent, strontium takes its effect on osteoblasts at multilevel through multiple pathways, representing a good example in revealing and exploring anabolic mechanisms. The recognition of strontium effects on bone has led to its expected application in a variety of biomaterial scaffolds used in tissue engineering strategies aiming at bone repairing and regeneration. While summarizing the recent progress in these respects, this review also proposes the new approaches such as systems biology in order to reveal new insights in the pathology of osteoporosis as well as possible discovery of new therapies.
Collapse
|