1
|
Chen X, Wei Y, Li Z, Zhou C, Fan Y. Distinct role of Klotho in long bone and craniofacial bone: skeletal development, repair and regeneration. PeerJ 2024; 12:e18269. [PMID: 39465174 PMCID: PMC11505971 DOI: 10.7717/peerj.18269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/17/2024] [Indexed: 10/29/2024] Open
Abstract
Bone defects are highly prevalent diseases caused by trauma, tumors, inflammation, congenital malformations and endocrine abnormalities. Ideally effective and side effect free approach to dealing with bone defects remains a clinical conundrum. Klotho is an important protein, which plays an essential role in regulating aging and mineral ion homeostasis. More recently, research revealed the function of Klotho in regulating skeleton development and regeneration. Klotho has been identified in mesenchymal stem cells, osteoblasts, osteocytes and osteoclasts in different skeleton regions. The specific function and regulatory mechanisms of Klotho in long bone and craniofacial bone vary due to their different embryonic development, ossification and cell types, which remain unclear and without conclusion. Moreover, studies have confirmed that Klotho is a multifunctional protein that can inhibit inflammation, resist cancer and regulate the endocrine system, which may further accentuate the potential of Klotho to be the ideal molecule in inducing bone restoration clinically. Besides, as an endogenous protein, Klotho has a promising potential for clinical therapy without side effects. In the current review, we summarized the specific function of Klotho in long bone and craniofacial skeleton from phenotype to cellular alternation and signaling pathway. Moreover, we illustrated the possible future clinical application for Klotho. Further research on Klotho might help to solve the existing clinical difficulties in bone healing and increase the life quality of patients with bone injury and the elderly.
Collapse
Affiliation(s)
- Xinyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yali Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zucen Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Papatheodorou V, Gerodimos C, Dimitrakopoulos A, Lada E, Tektonidou MG, Germenis A, Sfikakis PP, Laskari K. TNFRSF11A variants contribute to systemic autoinflammatory diseases: A case series of 12 patients. Semin Arthritis Rheum 2024; 68:152505. [PMID: 39003954 DOI: 10.1016/j.semarthrit.2024.152505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/07/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Limited evidence suggests that variants in TNFRSF11A gene, encoding RANK, may contribute to systemic autoinflammatory disease (SAID). AIM/METHODS To estimate the prevalence of TNFRSF11A variants in a cohort of patients with SAIDs screened for 26 related genes and describe the disease phenotypic expression. RESULTS A total of 12 out of 167 patients, 7 males, aged (median) 38 years at disease onset, yielded at least one TNFRSF11A rare variant. All patients carried a coexisting variant in at least one other SAID-related gene, most frequently MEFV (6 patients), but also TNFRSF1A, NOD2, NLRP3, NLRP7, MVK, IL36RN, RBCK1, PLCG2 and PSMB8. SAID episodes lasting (median) 9 days manifested with high grade fever (91%), myalgias (75%), malaise (67%), serositis (58%), arthralgias/arthritis (58%), gastrointestinal involvement (33%), and rash (25%), and responded to corticosteroids. The most common initial clinical diagnosis was TNF-associated periodic fever syndrome (TRAPS), which was, however, confirmed, in only one patient. The emergence of MEFV variations supported the diagnosis of atypical Familial Mediterranean Fever in two cases, whereas the diagnosis of Yao syndrome was speculated in two patients with NOD2 variants. The presence of atypical disease and the inability of defining diagnosis in the remaining 7 patients, supported the possible involvement of TNFRSF11A variants in the phenotypic expression of SAIDs. CONCLUSION TNFRSF11A variants, occurring in 7% of SAID patients always in combination with other SAID-related gene variants, contribute to the development of an autoinflammatory syndrome resembling to TRAPS. Additional studies to confirm novel pathogenic SAID pathways are clearly warranted.
Collapse
Affiliation(s)
- Vasileios Papatheodorou
- Joint Academic Rheumatology Program, First Department of Propaedeutic and Internal Medicine, National and Kapodistrian University of Athens, Greece
| | | | | | | | - Maria G Tektonidou
- Joint Academic Rheumatology Program, First Department of Propaedeutic and Internal Medicine, National and Kapodistrian University of Athens, Greece
| | - Anastasios Germenis
- Department of Immunology and Histocompatibility, School of Medicine, University of Thessaly, Larissa, Greece
| | - Petros P Sfikakis
- Joint Academic Rheumatology Program, First Department of Propaedeutic and Internal Medicine, National and Kapodistrian University of Athens, Greece
| | - Katerina Laskari
- Joint Academic Rheumatology Program, First Department of Propaedeutic and Internal Medicine, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
3
|
Yamada S, Chea C, Furusho H, Oda K, Shiba F, Tanimoto K, Tate SI, Miyauchi M, Takata T. Effects of novel lactoferrin peptides on LPS-induced alveolar bone destruction in a rat model. Chem Biol Drug Des 2024; 104:e14574. [PMID: 38958121 DOI: 10.1111/cbdd.14574] [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: 12/02/2023] [Revised: 04/14/2024] [Accepted: 04/23/2024] [Indexed: 07/04/2024]
Abstract
To develop novel bovine lactoferrin (bLF) peptides targeting bLF-tumour necrosis factor (TNF) receptor-associated factor 6 (TRAF6) binding sites, we identified two peptides that could target bLF-TRAF6 binding sites using structural analysis. Moreover, another peptide that could bind to the TRAF6 dimerization area was selected from the bLF sequence. The effects of each peptide on cytokine expression in lipopolysaccharide (LPS)-stimulated osteoblasts (ST2) and on osteoclastogenesis were examined using an LPS-treated co-culture of primary bone marrow cells (BMCs) with ST2 cells and a single culture of osteoclast precursor cells (RAW-D) treated with soluble receptor activator of NF-κB ligand. Finally, the effectiveness of these peptides against LPS-induced alveolar bone destruction was assessed. Two of the three peptides significantly suppressed LPS-induced TNF-α and interleukin-1β expression in ST2 cells. Additionally, these peptides inhibited and reversed LPS-induced receptor activator of NF-κB ligand (RANKL) upregulation and osteoprotegerin (OPG) downregulation, respectively. Furthermore, both peptides significantly reduced LPS-induced osteoclastogenesis in the BMC-ST2 co-culture and RANKL-induced osteoclastogenesis in RAW-D cells. In vivo, topical application of these peptides significantly reduced the osteoclast number by downregulating RANKL and upregulating OPG in the periodontal ligament. It is indicated that the novel bLF peptides can be used to treat periodontitis-associated bone destruction.
Collapse
Affiliation(s)
- Sakura Yamada
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Orthodontics and Craniofacial Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Chanbora Chea
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Faculty of Dentistry, University of Puthisastra, Phnom Penh, Cambodia
| | - Hisako Furusho
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kanae Oda
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Orthodontics and Craniofacial Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Fumie Shiba
- Collaborative Research Laboratory of Oral Inflammation Regulation, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Kotaro Tanimoto
- Department of Orthodontics and Craniofacial Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shin-Ichi Tate
- Laboratory of Molecular Biophysics, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
- SKCM2, Hiroshima University, Hiroshima, Japan
| | - Mutsumi Miyauchi
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Collaborative Research Laboratory of Oral Inflammation Regulation, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Takashi Takata
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Shunan University, Yamaguchi, Japan
| |
Collapse
|
4
|
Kim TJ, MacElroy AS, Defreitas A, Shenker BJ, Boesze-Battaglia K. A Synthetic Small Molecule, LGM2605: A Promising Modulator of Increased Pro-Inflammatory Cytokine and Osteoclast Differentiation by Aggregatibacter actinomycetemcomitans Cytolethal Distending Toxin. Dent J (Basel) 2024; 12:195. [PMID: 39056982 PMCID: PMC11276599 DOI: 10.3390/dj12070195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Our research explores the interplay between Aggregatibacter actinomycetemcomitans (Aa) cytolethal distending toxin (Cdt) and the host's inflammatory response in molar/incisor pattern periodontitis (MIPP). Cdt disrupts phosphatidylinositol-3,4,5-triphosphate (PIP3) signaling, influencing cytokine expression through canonical and non-canonical inflammasome activation as well as nuclear factor-κB (NF-κB) activation, leading to inflammation in MIPP. THP-1 differentiated macrophages (TDMs) exposed to Cdt exhibited an upregulation of pro-inflammatory genes and subsequent cytokine release. We analyzed the ability of a small molecule therapeutic, LGM2605, known for its anti-inflammatory properties, to reduce pro-inflammatory gene expression and cytokine release in Cdt-exposed and Aa-inoculated TDMs. LGM2605's mechanism of action involves inhibiting NF-κB while activating the Nrf2-transcription factor and antioxidants. Herein, we show that this small molecule therapeutic mitigates Cdt-induced pro-inflammatory cytokine expression and secretion. Our study also further defines Cdt's impact on osteoclast differentiation and maturation in MIPP. Cdt promotes increased TRAP+ cells, indicating heightened osteoclast differentiation, specific to Cdt's phosphatase activity. Cathepsin K levels rise during this process, reflecting changes in TRAP distribution between control and Cdt-treated cells. Exploring LGM2605's effect on Cdt-induced osteoclast differentiation and maturation, we found TRAP+ cells significantly reduced with LGM2605 treatment compared to Cdt alone. Upon LGM2605 treatment, immunocytochemistry revealed a decreased TRAP intensity and number of multinucleated cells. Moreover, immunoblotting showed reduced TRAP and cathepsin K levels, suggesting LGM2605's potential to curb osteoclast differentiation and maturation by modulating inflammatory cytokines, possibly involving Nrf2 activation. In summary, our research reveals the intricate connections between Cdt, pro-inflammatory cytokines, and osteoclast differentiation, offering novel therapeutic possibilities for managing these conditions.
Collapse
Affiliation(s)
- Taewan J. Kim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.S.M.); (A.D.); (B.J.S.)
| | - Andrew S. MacElroy
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.S.M.); (A.D.); (B.J.S.)
| | - Aleena Defreitas
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.S.M.); (A.D.); (B.J.S.)
| | - Bruce J. Shenker
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.S.M.); (A.D.); (B.J.S.)
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.S.M.); (A.D.); (B.J.S.)
| |
Collapse
|
5
|
Cao S, Li Y, Song R, Meng X, Fuchs M, Liang C, Kachler K, Meng X, Wen J, Schlötzer-Schrehardt U, Taudte V, Gessner A, Kunz M, Schleicher U, Zaiss MM, Kastbom A, Chen X, Schett G, Bozec A. L-arginine metabolism inhibits arthritis and inflammatory bone loss. Ann Rheum Dis 2024; 83:72-87. [PMID: 37775153 PMCID: PMC10803985 DOI: 10.1136/ard-2022-223626] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 08/29/2023] [Indexed: 10/01/2023]
Abstract
OBJECTIVES To investigate the effect of the L-arginine metabolism on arthritis and inflammation-mediated bone loss. METHODS L-arginine was applied to three arthritis models (collagen-induced arthritis, serum-induced arthritis and human TNF transgenic mice). Inflammation was assessed clinically and histologically, while bone changes were quantified by μCT and histomorphometry. In vitro, effects of L-arginine on osteoclast differentiation were analysed by RNA-seq and mass spectrometry (MS). Seahorse, Single Cell ENergetIc metabolism by profilIng Translation inHibition and transmission electron microscopy were used for detecting metabolic changes in osteoclasts. Moreover, arginine-associated metabolites were measured in the serum of rheumatoid arthritis (RA) and pre-RA patients. RESULTS L-arginine inhibited arthritis and bone loss in all three models and directly blocked TNFα-induced murine and human osteoclastogenesis. RNA-seq and MS analyses indicated that L-arginine switched glycolysis to oxidative phosphorylation in inflammatory osteoclasts leading to increased ATP production, purine metabolism and elevated inosine and hypoxanthine levels. Adenosine deaminase inhibitors blocking inosine and hypoxanthine production abolished the inhibition of L-arginine on osteoclastogenesis in vitro and in vivo. Altered arginine levels were also found in RA and pre-RA patients. CONCLUSION Our study demonstrated that L-arginine ameliorates arthritis and bone erosion through metabolic reprogramming and perturbation of purine metabolism in osteoclasts.
Collapse
Affiliation(s)
- Shan Cao
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
- Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yixuan Li
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
- Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Song
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
- Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xianyi Meng
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
| | - Maximilian Fuchs
- Chair of Medical Informatics, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Chunguang Liang
- Chair of Medical Informatics, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Bioinformatics, Biocenter, University of Würzburg Am Hubland, Würzburg, Germany
| | - Katerina Kachler
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
| | - Xinyu Meng
- Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinming Wen
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Verena Taudte
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Core Facility for Metabolomics, Department of Medicine, Philipps University of Marburg, Marburg, Germany
| | - Arne Gessner
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Meik Kunz
- Chair of Medical Informatics, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Ulrike Schleicher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mario M Zaiss
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
| | - Alf Kastbom
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Xiaoxiang Chen
- Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
| | - Aline Bozec
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Shanghai, Germany
| |
Collapse
|
6
|
Khocht A, Bellinger D, Lenoir L, Irani C, Fraser G. Obesity Is Associated with a Weakened Gingival Inflammatory Cytokine Response. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2089. [PMID: 38138192 PMCID: PMC10744834 DOI: 10.3390/medicina59122089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023]
Abstract
Background and Objectives: An obesity-related elevated body mass index (BMI) across life is associated with chronic low-grade inflammation and increased levels of C-reactive protein (CRP) in blood. CRP is a marker and promoter of inflammation. The objectives of this study were to examine the effect of obesity on the relationship between peripheral and gingival CRP levels and to examine the effects of gingival CRP levels on gingival fluid inflammatory cytokines in periodontitis-resistant obese individuals. Materials and Methods: Thirty-nine participants in good periodontal health were recruited. Twenty patients were classified as lean and nineteen as obese based on their BMI levels. A thorough periodontal assessment was carried out. Gingival crevicular fluid (GCF) and blood samples were collected. Both GCF and blood samples were analyzed for interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8), tumor necrosis factor-α (TNF-α), interleukin-10 (IL-10), interleukin-17A (IL-17A), and CRP. Results: GCF CRP levels were significantly higher in the obese than in the lean individuals. No statistically significant differences were noted between the two groups in either GCF or blood in terms of any of the inflammatory cytokine levels. IL-17A was not detected in the GCF of most subjects in both groups. GCF CRP levels were positively associated with blood CRP levels, and the association tended to be stronger in the obese individuals. GCF CRP showed no associations with GCF IL-10 in both groups. Although GCF CRP levels were positively associated with multiple GCF inflammatory cytokines (e.g., IL-1β, IL-6, IL-8, and TNF-α) in all subjects, the associations tended to be weaker in the obese individuals (e.g., IL-1β, IL-6, and TNF-α). Furthermore, the levels of the GCF inflammatory cytokines IL-6 and TNF-α were decreased in the obese individuals. Conclusions: Obesity unfavorably influences the relationship between blood and GCF CRP levels and promotes increased CRP levels in GCF. Collectively, the findings suggest a weakened inflammatory cytokine response in the gingival tissues of obese individuals.
Collapse
Affiliation(s)
- Ahmed Khocht
- Department of Periodontics, School of Dentistry, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Denise Bellinger
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Leticia Lenoir
- Department of Periodontics, School of Dentistry, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Crissy Irani
- Institute for Community Partnerships, Loma Linda University Health, Loma Linda, CA 92354, USA;
| | - Gary Fraser
- Department of Preventive Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
| |
Collapse
|
7
|
Kondegowda NG, Filipowska J, Do JS, Leon-Rivera N, Li R, Hampton R, Ogyaadu S, Levister C, Penninger JM, Reijonen H, Levy CJ, Vasavada RC. RANKL/RANK is required for cytokine-induced beta cell death; osteoprotegerin, a RANKL inhibitor, reverses rodent type 1 diabetes. SCIENCE ADVANCES 2023; 9:eadf5238. [PMID: 37910614 PMCID: PMC10619938 DOI: 10.1126/sciadv.adf5238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 09/29/2023] [Indexed: 11/03/2023]
Abstract
Treatment for type 1 diabetes (T1D) requires stimulation of functional β cell regeneration and survival under stress. Previously, we showed that inhibition of the RANKL/RANK [receptor activator of nuclear factor kappa Β (NF-κB) ligand] pathway, by osteoprotegerin and the anti-osteoporotic drug denosumab, induces rodent and human β cell proliferation. We demonstrate that the RANK pathway mediates cytokine-induced rodent and human β cell death through RANK-TRAF6 interaction and induction of NF-κB activation. Osteoprotegerin and denosumab protected β cells against this cytotoxicity. In human immune cells, osteoprotegerin and denosumab reduce proinflammatory cytokines in activated T-cells by inhibiting RANKL-induced activation of monocytes. In vivo, osteoprotegerin reversed recent-onset T1D in nonobese diabetic/Ltj mice, reduced insulitis, improved glucose homeostasis, and increased plasma insulin, β cell proliferation, and mass in these mice. Serum from T1D subjects induced human β cell death and dysfunction, but not α cell death. Osteoprotegerin and denosumab reduced T1D serum-induced β cell cytotoxicity and dysfunction. Inhibiting RANKL/RANK could have therapeutic potential.
Collapse
Affiliation(s)
- Nagesha Guthalu Kondegowda
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joanna Filipowska
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jeong-su Do
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Nancy Leon-Rivera
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Rosemary Li
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rollie Hampton
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Selassie Ogyaadu
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Endocrinology and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Camilla Levister
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Endocrinology and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Josef M. Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna 1030, Austria
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Helena Reijonen
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Carol J. Levy
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Endocrinology and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rupangi C. Vasavada
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
8
|
Rajeshwari HRS, Kishen A. Periodontal Fibroblasts-Macrophage Crosstalk in External Inflammatory Root Resorption. J Endod 2023; 49:1145-1153.e3. [PMID: 37268291 DOI: 10.1016/j.joen.2023.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/04/2023]
Abstract
INTRODUCTION This study aimed to understand the influence of periodontal fibroblasts (PDLFs) on clastic differentiation of macrophages (Mφ) in different resorptive environments. METHODS PDLF-Mφ direct coculture (juxtacrine) was seeded on dentin, cementum, and polystyrene with/without lipopolysaccharide, macrophage colony-stimulating factor, and receptor activator of nuclear factor kappa beta ligand for 7 and 14 days and stained for tartrate-resistant acid phosphatase (TRAP) activity. PDLF-Mφ cocultured on polystyrene were immunostained for CD80, CD206, NFATc1, STAT6, and periostin, and cell culture supernatants were assessed for cytokines on days 2 and 7. Mφ grown in conditioned media of PDLFs (paracrine) and Mφ monoculture were used as controls. Data was analyzed using Student t test and one-way analysis of variance with the Tukey multiple comparisons test (P < .05). RESULTS PDLF-Mφ coculture showed a higher number of TRAP-positive multinucleated cells than Mφ monoculture on dentin and polystyrene. No TRAP-positive multinucleated cells were observed in paracrine and cementum. The expression of CD80 and CD206 in PDLF-Mφ was similar at day 2, whereas CD206 was greater than CD80 at day 7. The expression of STAT6 was greater than NFATc1 at both days 2 and 7 (P < .05). Periostin expression in the presence of the lipopolysaccharide, macrophage colony-stimulating factor, and receptor activator of nuclear factor kappa beta ligand combination was down-regulated in PDLF monoculture, whereas it was up-regulated in PDLF-Mφ coculture. The cytokine profile of PDLF-Mφ on day 2 was predominated by interleukin (IL)-1β, tumor necrosis factor alpha, and MMP9 and MMP2 on day 7. IL-6 and IL-8 showed steady expression at both days 2 and 7. CONCLUSIONS The study highlights the juxtacrine effect of PDLFs on the clastic differentiation of Mφ with a difference in clastic activity between dentin and cementum. The study also emphasizes the temporal effect of tumor necrosis factor alpha, MMP2, MMP9, and IL-1β on intercellular crosstalk in resorptive environments.
Collapse
Affiliation(s)
| | - Anil Kishen
- The Kishen Lab, Dental Research Institute, University of Toronto, Toronto, Ontario, Canada; Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada; School of Graduate Studies, University of Toronto, Toronto, Ontario, Canada; Department of Dentistry, Mount Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
9
|
Dermatologic Manifestations of Noninflammasome-Mediated Autoinflammatory Diseases. JID INNOVATIONS 2023; 3:100176. [PMID: 36876221 PMCID: PMC9982332 DOI: 10.1016/j.xjidi.2022.100176] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022] Open
Abstract
Autoinflammatory diseases (AIDs) arise from disturbances that alter interactions of immune cells and tissues. They give rise to prominent (auto)inflammation in the absence of aberrant autoantibodies and/or autoreactive T cells. AIDs that are predominantly caused by changes in the inflammasome pathways, such as the NLRP3- or pyrin-associated inflammasome, have gained substantial attention over the last years. However, AIDs resulting primarily from other changes in the defense system of the innate immune system are less well-studied. These noninflammasome-mediated AIDs relate to, for example, disturbance in the TNF or IFN signaling pathways or aberrations in genes affecting the IL-1RA. The spectrum of clinical signs and symptoms of these conditions is vast. Thus, recognizing early cutaneous signs constitutes an important step in differential diagnoses for dermatologists and other physicians. This review provides an overview of the pathogenesis, clinical presentation, and available treatment options highlighting dermatologic aspects of noninflammasome-mediated AIDs.
Collapse
Key Words
- AID, autoinflammatory disease
- ANCA, antineutrophil cytoplasmic antibody
- AOSD, adult-onset Still disease
- BASDAI, Bath Ankylosing Spondylitis Activity Index
- CANDLE, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature
- CAPS, cryopyrin-associated periodic syndrome
- CRD, cysteine-rich domain
- DIRA, deficiency of IL-1RA
- DITRA, deficiency of IL-36RA
- ER, endoplasmic reticulum
- ESR, erythrocyte sedimentation rate
- FMF, familial Mediterranean fever
- M-CSF, macrophage colony-stimulating factor
- MAS, macrophage activation syndrome
- NET, neutrophil extracellular trap
- NOS, nitrous oxide
- NSAID, nonsteroidal anti-inflammatory drug
- NUD, neutrophilic urticarial dermatosis
- PFAPA, periodic fever, aphthous stomatitis, pharyngitis, and adenitis
- PKR, protein kinase R
- PRAAS, proteosome-associated autoinflammatory disease
- SAPHO, synovitis, acne, pustulosis, hyperostosis, osteitis syndrome
- SAVI, STING-associated vasculopathy with onset in infancy
- STAT, signal transducer and activator of transcription
- SchS, Schnitzler syndrome
- TNFR, TNF receptor
- TRAPS, TNF receptor‒associated autoinflammatory disease
- Th17, T helper 17
- VAS, Visual Analog Scale
- sTNFR, soluble TNF receptor
Collapse
|
10
|
Sun B, Sun Y, Han X, Ma Q, Meng Q. Leucine supplementation alleviates immune and antioxidant function damage in adult rats induced by early weaning. J Nutr 2023; 153:1607-1617. [PMID: 37004874 DOI: 10.1016/j.tjnut.2023.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Early weaning (EW) can lead to stress and destroy intestinal integrity. Leucine has functional diversity in antioxidant, immune, and metabolic regulation. OBJECTIVE This study aims to explore a lifelong impact of early weaning on intestinal, immune and antioxidant functions of adult rats and the role of leucine supplementation in the alleviation of the damage caused by early weaning. METHODS In this 211-day study, 36 SD rat pups were divided into three groups: 21-day weaning normal group (C), 17-day early weaning group (E) and 17-day early weaning group with two-month leucine supplementation (EL). The content of amino acids in serum, immune and antioxidant indexes, intestinal morphology, liver transcriptomics, mRNA, and protein expression of signaling pathway were determined. RESULTS EW reduced the protein expression level of sIgA and GSH in jejunum, and increased the protein expression levels of IgA, IgM, and IL-17 in serum, and TNFα and IL-1β in jejunum. The impairment by EW was activated via NF-κB signal pathway. In terms of antioxidation, EW reduced the level of GSH in jejunum. After leucine supplementation, the damage induced by EW was partially repaired. CONCLUSIONS EW causes long-term damage to the intestinal barrier function, immunity, apoptosis factor, and antioxidant function of rats and leucine supplementation could alleviate the impairment, suggesting possible approach to EW.
Collapse
Affiliation(s)
- Bo Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.
| | - Yuchen Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.
| | - Xuesong Han
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.
| | - Qingquan Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.
| | - Qingwei Meng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
11
|
Chen WA, Dou Y, Fletcher HM, Boskovic DS. Local and Systemic Effects of Porphyromonas gingivalis Infection. Microorganisms 2023; 11:470. [PMID: 36838435 PMCID: PMC9963840 DOI: 10.3390/microorganisms11020470] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023] Open
Abstract
Porphyromonas gingivalis, a gram-negative anaerobe, is a leading etiological agent in periodontitis. This infectious pathogen can induce a dysbiotic, proinflammatory state within the oral cavity by disrupting commensal interactions between the host and oral microbiota. It is advantageous for P. gingivalis to avoid complete host immunosuppression, as inflammation-induced tissue damage provides essential nutrients necessary for robust bacterial proliferation. In this context, P. gingivalis can gain access to the systemic circulation, where it can promote a prothrombotic state. P. gingivalis expresses a number of virulence factors, which aid this pathogen toward infection of a variety of host cells, evasion of detection by the host immune system, subversion of the host immune responses, and activation of several humoral and cellular hemostatic factors.
Collapse
Affiliation(s)
- William A. Chen
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Yuetan Dou
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Hansel M. Fletcher
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Danilo S. Boskovic
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| |
Collapse
|
12
|
Body Composition in Patients with Follicular Lymphoma: Asso-Ciations between Changes in Radiomic Parameters in Patients Treated with R-CHOP-like and R-B Regimens: LyRa 01F. Cancers (Basel) 2023; 15:cancers15040999. [PMID: 36831345 PMCID: PMC9954461 DOI: 10.3390/cancers15040999] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/09/2023] Open
Abstract
In patients with follicular lymphoma (FL), therapeutic advances have led to improved survival, and within this framework, it is important to identify treatment strategies offering a better quality of life. Using (18)F-fluorodeoxyglucose positron emission tomography/computed tomography (PET/CT), in patients treated with R-CHOP-like or R-Bendamustine regimens, we assessed changes in the bone mineral density (BMD), musculoskeletal index (SMI), visceral adipose tissue (VAT), and subcutaneous adipose tissue (SAT) at disease onset and at the end of therapy. We evaluated whether the high-steroid regimen could lead to more significant radiological changes than those induced by the steroid-free regimen and whether a low BMD at disease onset is an unfavorable prognostic index. Seventy-nine patients between 60 and 80 years old with a new diagnosis of FL were included in the study. Evaluation of Delta values (pre- and post-therapy mean values) in the two immunochemotherapy regimens showed differences in radiomic parameters within the two patient cohorts. The R-CHOP-like regimen was associated with a significant reduction in BMD, an increase in SAT and VAT, and a reduction in skeletal muscle density (SMD) and SMI. Moreover, patients with high FLIPI showed a BMD below the cut-off value. This study represents the first study demonstrating a prognostic correlation between FLIPI and low BMD.
Collapse
|
13
|
Assadiasl S, Rajabinejad M, Soleimanifar N, Makiyan F, Azizi E, Rezaiemanesh A, Nicknam MH. MicroRNAs-mediated regulation pathways in rheumatic diseases. Inflammopharmacology 2023; 31:129-144. [PMID: 36469219 DOI: 10.1007/s10787-022-01097-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 10/29/2022] [Indexed: 12/09/2022]
Abstract
Rheumatoid arthritis (RA) and ankylosing spondylitis (AS) are two common rheumatic disorders marked by persistent inflammatory joint disease. Patients with RA have osteodestructive symptoms, but those with AS have osteoproliferative manifestations. Ligaments, joints, tendons, bones, and muscles are all affected by rheumatic disorders. In recent years, many epigenetic factors contributing to the pathogenesis of rheumatoid disorders have been studied. MicroRNAs (miRNAs) are small, non-coding RNA molecules implicated as potential therapeutic targets or biomarkers in rheumatic diseases. MiRNAs play a critical role in the modulation of bone homeostasis and joint remodeling by controlling fibroblast-like synoviocytes (FLSs), chondrocytes, and osteocytes. Several miRNAs have been shown to be dysregulated in rheumatic diseases, including miR-10a, 16, 17, 18a, 19, 20a, 21, 27a, 29a, 34a, 103a, 125b, 132, 137, 143, 145, 146a, 155, 192, 203, 221, 222, 301a, 346, and 548a.The major molecular pathways governed by miRNAs in these cells are Wnt, bone-morphogenic protein (BMP), nuclear factor (NF)-κB, receptor activator of NF-κB (RANK)-RANK ligand (RANKL), and macrophage colony-stimulating factor (M-CSF) receptor pathway. This review aimed to provide an overview of the most important signaling pathways controlled by miRNAs in rheumatic diseases.
Collapse
Affiliation(s)
- Sara Assadiasl
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Misagh Rajabinejad
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Narjes Soleimanifar
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Makiyan
- Division of Nanobiotechnology, Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Esfandiar Azizi
- Department of Immunology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Alireza Rezaiemanesh
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Daneshgah Street, Shahid Shiroudi Boulevard, PO-Box: 6714869914, Bākhtarān, Iran.
| | | |
Collapse
|
14
|
Zhou P, Zheng T, Zhao B. Cytokine-mediated immunomodulation of osteoclastogenesis. Bone 2022; 164:116540. [PMID: 36031187 PMCID: PMC10657632 DOI: 10.1016/j.bone.2022.116540] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022]
Abstract
Cytokines are an important set of proteins regulating bone homeostasis. In inflammation induced bone resorption, cytokines, such as RANKL, TNF-α, M-CSF, are indispensable for the differentiation and activation of resorption-driving osteoclasts, the process we know as osteoclastogenesis. On the other hand, immune system produces a number of regulatory cytokines, including IL-4, IL-10 and IFNs, and limits excessive activation of osteoclastogenesis and bone loss during inflammation. These unique properties make cytokines powerful targets as rheostat to maintain bone homeostasis and for potential immunotherapies of inflammatory bone diseases. In this review, we summarize recent advances in cytokine-mediated regulation of osteoclastogenesis and provide insights of potential translational impact of bench-side research into clinical treatment of bone disease.
Collapse
Affiliation(s)
- Pengcheng Zhou
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China; Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
| | - Ting Zheng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA; Graduate Program in Biochemistry, Cell and Molecular Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| |
Collapse
|
15
|
Decoding the transcriptome of calcified atherosclerotic plaque at single-cell resolution. Commun Biol 2022; 5:1084. [PMID: 36224302 PMCID: PMC9556750 DOI: 10.1038/s42003-022-04056-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 09/30/2022] [Indexed: 11/30/2022] Open
Abstract
Atherogenesis involves an interplay of inflammation, tissue remodeling and cellular transdifferentiation (CTD), making it especially difficult to precisely delineate its pathophysiology. Here we use single-cell RNA sequencing and systems-biology approaches to analyze the transcriptional profiles of vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) in calcified atherosclerotic core (AC) plaques and patient-matched proximal adjacent (PA) portions of carotid artery tissue from patients undergoing carotid endarterectomy. Our results reveal an anatomic distinction whereby PA cells express inflammatory mediators, while cells expressing matrix-secreting genes occupy a majority of the AC region. Systems biology analysis indicates that inflammation in PA ECs and VSMCs may be driven by TNFa signaling. Furthermore, we identify POSTN, SPP1 and IBSP in AC VSMCs, and ITLN1, SCX and S100A4 in AC ECs as possible candidate drivers of CTD in the atherosclerotic core. These results establish an anatomic framework for atherogenesis which forms the basis for exploration of a site-specific strategy for disruption of disease progression. Single-cell RNA sequencing and systems biology are used to profile the human vascular cell populations in calcified atherosclerotic core plaques from carotid endarterectomy samples, showing an anatomic distinction between gene expression of inflammatory versus matrix-secreting factors.
Collapse
|
16
|
Nguyen CT, Furuya H, Das D, Marusina AI, Merleev AA, Ravindran R, Jalali Z, Khan IH, Maverakis E, Adamopoulos IE. Peripheral γδ T Cells Regulate Neutrophil Expansion and Recruitment in Experimental Psoriatic Arthritis. Arthritis Rheumatol 2022; 74:1524-1534. [PMID: 35320625 PMCID: PMC9427669 DOI: 10.1002/art.42124] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/01/2022] [Accepted: 03/17/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE This study was undertaken to identify the mechanistic role of γδ T cells in the pathogenesis of experimental psoriatic arthritis (PsA). METHODS In this study, we performed interleukin-23 (IL-23) gene transfer in wild-type (WT) and T cell receptor δ-deficient (TCRδ-/- ) mice and conducted tissue phenotyping in the joint, skin, and nails to characterize the inflammatory infiltrate. We further performed detailed flow cytometry, immunofluorescence staining, RNA sequencing, T cell repertoire analysis, and in vitro T cell polarization assays to identify regulatory mechanisms of γδ T cells. RESULTS We demonstrated that γδ T cells support systemic granulopoiesis, which is critical for murine PsA-like pathology. Briefly, γδ T cell ablation inhibited the expression of neutrophil chemokines CXCL1 and CXCL2 and neutrophil CD11b+Ly6G+ accumulation in the aforementioned PsA-related tissues. Although significantly reduced expression of granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-17A was detected systemically in TCRδ-/- mice, no GM-CSF+/IL-17A+ γδ T cells were detected locally in the inflamed skin or bone marrow in WT mice. Our data showed that nonresident γδ T cells regulate the expansion of an CD11b+Ly6G+ neutrophil population and their recruitment to joint and skin tissues, where they develop hallmark pathologic features of human PsA. CONCLUSION Our findings do not support the notion that tissue-resident γδ T cells initiate the disease but demonstrate a novel role of γδ T cells in neutrophil regulation that can be exploited therapeutically in PsA patients.
Collapse
Affiliation(s)
- Cuong Thach Nguyen
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis
| | - Hiroki Furuya
- Department of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| | - Dayasagar Das
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis
| | - Alina I Marusina
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Alexander A Merleev
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Resmi Ravindran
- Department of Pathology and Laboratory Medicine, University of California at Davis, USA
| | - Zahra Jalali
- Department of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| | - Imran H. Khan
- Department of Pathology and Laboratory Medicine, University of California at Davis, USA
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Iannis E. Adamopoulos
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis
- Department of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| |
Collapse
|
17
|
Pesce B, Ribeiro CH, Larrondo M, Ramos V, Soto L, Catalán D, Aguillón JC. TNF-α Affects Signature Cytokines of Th1 and Th17 T Cell Subsets through Differential Actions on TNFR1 and TNFR2. Int J Mol Sci 2022; 23:ijms23169306. [PMID: 36012570 PMCID: PMC9408897 DOI: 10.3390/ijms23169306] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor necrosis factor (TNF)-α is a pleiotropic cytokine implicated in the etiology of several autoimmune diseases, including rheumatoid arthritis (RA). TNF-α regulates diverse effector functions through the activation of TNF-α receptor (TNFR)1 and TNFR2. Although the detrimental role of this cytokine has been addressed in distinct disease settings, the effects of TNF-α on cytokine production by isolated CD4+ T helper type 1 (Th1) and Th17 cells, two T cell subpopulations that contribute to the pathogenesis of RA, have not been completely elucidated. Here, we show that TNF-α promotes a reduction and expansion in the frequency of both T cell subsets producing IFN-γ and IL-17, respectively. Selective blockade of TNFR1 or TNFR2 on Th1 and Th17 cells revealed that TNFR2 mediates the decrease in IFN-γ production, while signaling through both receptors augments IL-17 production. We also demonstrate that Th1, but not Th17 cells from RA patients present lower levels of TNFR1 compared to healthy controls, whereas TNFR2 expression on both T cell types is similar between patients and controls. Since TNF-α receptors levels in RA patients are not significantly changed by the therapeutic blockade of TNF-α, we propose that targeting TNFR2 may represent an alternative strategy to normalize the levels of key cytokines that contribute to RA pathogenesis.
Collapse
Affiliation(s)
- Bárbara Pesce
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Laboratorio MED.UCHILE-FACS, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Carolina H. Ribeiro
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Milton Larrondo
- Hospital Clínico, Universidad de Chile, Santiago 8380453, Chile
| | - Verónica Ramos
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Hospital Clínico, Universidad de Chile, Santiago 8380453, Chile
| | - Lilian Soto
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Hospital Clínico, Universidad de Chile, Santiago 8380453, Chile
| | - Diego Catalán
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Correspondence: (D.C.); (J.C.A.)
| | - Juan Carlos Aguillón
- Immune Regulation and Tolerance Research Group (IRTGroup), Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Correspondence: (D.C.); (J.C.A.)
| |
Collapse
|
18
|
Concerted regulation of OPG/RANKL/ NF‑κB/MMP-13 trajectories contribute to ameliorative capability of prodigiosin and/or low dose γ-radiation against adjuvant- induced arthritis in rats. Int Immunopharmacol 2022; 111:109068. [PMID: 35944459 DOI: 10.1016/j.intimp.2022.109068] [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: 05/27/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prodigiosin (PDG) is a microbial red dye with antioxidant and anti-inflammatory properties, although its effect on rheumatoid arthritis (RA) remains uncertain. Also, multiple doses of low dose γ- radiation (LDR) have been observed to be as a successful intervention for RA. Thus, the purpose of this study was to investigate the ameliorative potential of PDG and/or LDR on adjuvant-induced arthritis (AIA) in rats. METHODS The anti-inflammatory and anti-arthritic effects of PDG and/or LDR were examined in vitro and in vivo, respectively. In the AIA model, the arthritic indexes, paw swelling degrees, body weight gain, and histopathological assessment in AIA rats were assayed. The impact of PDG (200 µg/kg; p.o) and/or LDR (0.5 Gy) on the levels of pro- and anti-inflammatory cytokines (IL-1β, TNF-α, IL-6, IL-18, IL-17A, and IL-10) as well as the regulation of osteoprotegrin (OPG)/ receptor activator of nuclear factor κB ligand (RANKL)/ nuclear factor-κB (NF-κB)/MMP-13 pathways was determined. Methotrexate (MTX; 0.05 mg/kg; twice/week, i.p) was administered concurrently as a standard anti-arthritic drug. RESULTS PDG and/or LDR markedly diminished the arthritic indexes, paw edema, weigh loss in AIA rats, alleviated the pathological alterations in joints, reduced the levels of pro-inflammatory cytokines IL-1β, TNF-α, IL-6, IL-18, IL-17A, and RANKL in serum and synovial tissues, while increasing anti-inflammatory cytokines IL-10 and OPG levels. Moreover, PDG and/or LDR down-regulated the expression of RANKL, NF-κBp65, MMP13, caspase-3, and decreased the RANKL/OPG ratio, whereas OPG and collagen II were enhanced in synovial tissues. CONCLUSION PDG and/or LDR exhibited obvious anti-RA activity on AIA.
Collapse
|
19
|
Behary P, Comninos AN. Bone Perspectives in Functional Hypothalamic Amenorrhoea: An Update and Future Avenues. Front Endocrinol (Lausanne) 2022; 13:923791. [PMID: 35795153 PMCID: PMC9251506 DOI: 10.3389/fendo.2022.923791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/11/2022] [Indexed: 01/22/2023] Open
Abstract
One of the most important and potentially long-lasting detrimental consequences of Functional Hypothalamic Amenorrhoea (FHA) is on skeletal homeostasis. Beyond oestrogen deficiency, FHA is associated with a cascade of additional neuro-endocrine and metabolic alterations, some adaptive, but which combine to disrupt skeletal homeostasis. Ultimately, this leads to a two-fold increased risk of fractures in women with FHA compared to healthy eumenorrhoeic women. Although the cornerstone of management of FHA-related bone loss remains recovery of menses via restoration of metabolic/psychological balance, there is rapidly developing evidence for hormonal manipulations (with a particular emphasis on route of administration) and other pharmacological treatments that can protect or improve skeletal homeostasis in FHA. In this mini-review, we provide an update on the pathophysiology, clinical management and future avenues in the field from a bone perspective.
Collapse
Affiliation(s)
- Preeshila Behary
- Endocrine Bone Unit, Imperial College Healthcare NHS Trust, London, United Kingdom
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Alexander N. Comninos
- Endocrine Bone Unit, Imperial College Healthcare NHS Trust, London, United Kingdom
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
20
|
Manual Therapy Facilitates Homeostatic Adaptation to Bone Microstructural Declines Induced by a Rat Model of Repetitive Forceful Task. Int J Mol Sci 2022; 23:ijms23126586. [PMID: 35743030 PMCID: PMC9223642 DOI: 10.3390/ijms23126586] [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: 05/11/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 01/27/2023] Open
Abstract
The effectiveness of manual therapy in reducing the catabolic effects of performing repetitive intensive force tasks on bones has not been reported. We examined if manual therapy could reduce radial bone microstructural declines in adult female Sprague–Dawley rats performing a 12-week high-repetition and high-force task, with or without simultaneous manual therapy to forelimbs. Additional rats were provided 6 weeks of rest after task cessation, with or without manual therapy. The control rats were untreated or received manual therapy for 12 weeks. The untreated TASK rats showed increased catabolic indices in the radius (decreased trabecular bone volume and numbers, increased osteoclasts in these trabeculae, and mid-diaphyseal cortical bone thinning) and increased serum CTX-1, TNF-α, and muscle macrophages. In contrast, the TASK rats receiving manual therapy showed increased radial bone anabolism (increased trabecular bone volume and osteoblast numbers, decreased osteoclast numbers, and increased mid-diaphyseal total area and periosteal perimeter) and increased serum TNF-α and muscle macrophages. Rest, with or without manual therapy, improved the trabecular thickness and mid-diaphyseal cortical bone attributes but not the mineral density. Thus, preventive manual therapy reduced the net radial bone catabolism by increasing osteogenesis, while rest, with or without manual therapy, was less effective.
Collapse
|
21
|
Baratchart E, Lo CH, Lynch CC, Basanta D. Integrated computational and in vivo models reveal Key Insights into macrophage behavior during bone healing. PLoS Comput Biol 2022; 18:e1009839. [PMID: 35559958 PMCID: PMC9106165 DOI: 10.1371/journal.pcbi.1009839] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/17/2022] [Indexed: 11/24/2022] Open
Abstract
Myeloid-derived monocyte and macrophages are key cells in the bone that contribute to remodeling and injury repair. However, their temporal polarization status and control of bone-resorbing osteoclasts and bone-forming osteoblasts responses is largely unknown. In this study, we focused on two aspects of monocyte/macrophage dynamics and polarization states over time: 1) the injury-triggered pro- and anti-inflammatory monocytes/macrophages temporal profiles, 2) the contributions of pro- versus anti-inflammatory monocytes/macrophages in coordinating healing response. Bone healing is a complex multicellular dynamic process. While traditional in vitro and in vivo experimentation may capture the behavior of select populations with high resolution, they cannot simultaneously track the behavior of multiple populations. To address this, we have used an integrated coupled ordinary differential equations (ODEs)-based framework describing multiple cellular species to in vivo bone injury data in order to identify and test various hypotheses regarding bone cell populations dynamics. Our approach allowed us to infer several biological insights including, but not limited to,: 1) anti-inflammatory macrophages are key for early osteoclast inhibition and pro-inflammatory macrophage suppression, 2) pro-inflammatory macrophages are involved in osteoclast bone resorptive activity, whereas osteoblasts promote osteoclast differentiation, 3) Pro-inflammatory monocytes/macrophages rise during two expansion waves, which can be explained by the anti-inflammatory macrophages-mediated inhibition phase between the two waves. In addition, we further tested the robustness of the mathematical model by comparing simulation results to an independent experimental dataset. Taken together, this novel comprehensive mathematical framework allowed us to identify biological mechanisms that best recapitulate bone injury data and that explain the coupled cellular population dynamics involved in the process. Furthermore, our hypothesis testing methodology could be used in other contexts to decipher mechanisms in complex multicellular processes. Myeloid-derived monocytes/macrophages are key cells for bone remodeling and injury repair. However, their temporal polarization status and control of bone-resorbing osteoclasts and bone-forming osteoblasts responses is largely unknown. In this study, we focused on two aspects of monocyte/macrophage population dynamics: 1) the injury-triggered pro- and anti-inflammatory monocytes/macrophages temporal profiles, 2) the contributions of pro- versus anti-inflammatory monocytes/macrophages in coordinating healing response. In order to test various hypotheses regarding bone cell populations dynamics, we have integrated a coupled ordinary differential equations-based framework describing multiple cellular species to in vivo bone injury data. Our approach allowed us to infer several biological insights including: 1) anti-inflammatory macrophages are key for early osteoclast inhibition and pro-inflammatory macrophage suppression, 2) pro-inflammatory macrophages are involved in osteoclast bone resorptive activity, whereas osteoblasts promote osteoclast differentiation, 3) Pro-inflammatory monocytes/macrophages rise during two expansion waves, which can be explained by the anti-inflammatory macrophages-mediated inhibition phase between the two waves. Taken together, this mathematical framework allowed us to identify biological mechanisms that recapitulate bone injury data and that explain the coupled cellular population dynamics involved in the process.
Collapse
Affiliation(s)
- Etienne Baratchart
- Integrated Mathematical Oncology Department, SRB4, Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Chen Hao Lo
- Cancer Biology Ph.D. Program, Department of Cell Biology Microbiology and Molecular Biology, University of South Florida, Tampa, Florida, United States of America
- Tumor Biology Department, SRB3, Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - Conor C. Lynch
- Cancer Biology Ph.D. Program, Department of Cell Biology Microbiology and Molecular Biology, University of South Florida, Tampa, Florida, United States of America
- * E-mail: (CL); (DB)
| | - David Basanta
- Integrated Mathematical Oncology Department, SRB4, Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
- * E-mail: (CL); (DB)
| |
Collapse
|
22
|
Fan Y, Cui C, Rosen CJ, Sato T, Xu R, Li P, Wei X, Bi R, Yuan Q, Zhou C. Klotho in Osx +-mesenchymal progenitors exerts pro-osteogenic and anti-inflammatory effects during mandibular alveolar bone formation and repair. Signal Transduct Target Ther 2022; 7:155. [PMID: 35538062 PMCID: PMC9090922 DOI: 10.1038/s41392-022-00957-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/03/2022] [Accepted: 03/06/2022] [Indexed: 02/08/2023] Open
Abstract
Maxillofacial bone defects are commonly seen in clinical practice. A clearer understanding of the regulatory network directing maxillofacial bone formation will promote the development of novel therapeutic approaches for bone regeneration. The fibroblast growth factor (FGF) signalling pathway is critical for the development of maxillofacial bone. Klotho, a type I transmembrane protein, is an important components of FGF receptor complexes. Recent studies have reported the presence of Klotho expression in bone. However, the role of Klotho in cranioskeletal development and repair remains unknown. Here, we use a genetic strategy to report that deletion of Klotho in Osx-positive mesenchymal progenitors leads to a significant reduction in osteogenesis under physiological and pathological conditions. Klotho-deficient mensenchymal progenitors also suppress osteoclastogenesis in vitro and in vivo. Under conditions of inflammation and trauma-induced bone loss, we find that Klotho exerts an inhibitory function on inflammation-induced TNFR signaling by attenuating Rankl expression. More importantly, we show for the first time that Klotho is present in human alveolar bone, with a distinct expression pattern under both normal and pathological conditions. In summary, our results identify the mechanism whereby Klotho expressed in Osx+-mensenchymal progenitors controls osteoblast differentiation and osteoclastogenesis during mandibular alveolar bone formation and repair. Klotho-mediated signaling is an important component of alveolar bone remodeling and regeneration. It may also be a target for future therapeutics.
Collapse
Affiliation(s)
- Yi Fan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Chen Cui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, 510055, Guangzhou, Guangdong, China
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Scarborough, ME, 04074, USA
| | - Tadatoshi Sato
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Ruoshi Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Peiran Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Xi Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Stomatology, 510055, Guangzhou, Guangdong, China
| | - Ruiye Bi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China.
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, Sichuan, China.
| |
Collapse
|
23
|
Carletti A, Cardoso C, Lobo-Arteaga J, Sales S, Juliao D, Ferreira I, Chainho P, Dionísio MA, Gaudêncio MJ, Afonso C, Lourenço H, Cancela ML, Bandarra NM, Gavaia PJ. Antioxidant and Anti-inflammatory Extracts From Sea Cucumbers and Tunicates Induce a Pro-osteogenic Effect in Zebrafish Larvae. Front Nutr 2022; 9:888360. [PMID: 35614979 PMCID: PMC9125325 DOI: 10.3389/fnut.2022.888360] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/31/2022] [Indexed: 12/24/2022] Open
Abstract
Bone metabolic disorders such as osteoporosis are characterized by the loss of mineral from the bone tissue leading to its structural weakening and increased susceptibility to fractures. A growing body of evidence suggests that inflammation and oxidative stress play an important role in the pathophysiological processes involved in the rise of these conditions. As the currently available therapeutic strategies are often characterized by toxic effects associated with their long-term use, natural antioxidants and anti-inflammatory compounds such as polyphenols promise to be a valuable alternative for the prevention and treatment of these disorders. In this scope, the marine environment is becoming an important source of bioactive compounds with potential pharmacological applications. Here, we explored the bioactive potential of three species of holothurians (Echinodermata) and four species of tunicates (Chordata) as sources of antioxidant and anti-inflammatory compounds with a particular focus on polyphenolic substances. Hydroethanolic and aqueous extracts were obtained from animals' biomass and screened for their content of polyphenols and their antioxidant and anti-inflammatory properties. Hydroethanolic fractions of three species of tunicates displayed high polyphenolic content associated with strong antioxidant potential and anti-inflammatory activity. Extracts were thereafter tested for their capacity to promote bone formation and mineralization by applying an assay that uses the developing operculum of zebrafish (Danio rerio) to assess the osteogenic activity of compounds. The same three hydroethanolic fractions from tunicates were characterized by a strong in vivo osteogenic activity, which positively correlated with their anti-inflammatory potential as measured by COX-2 inhibition. This study highlights the therapeutic potential of polyphenol-rich hydroethanolic extracts obtained from three species of tunicates as a substrate for the development of novel drugs for the treatment of bone disorders correlated to oxidative stress and inflammatory processes.
Collapse
Affiliation(s)
- Alessio Carletti
- Faculty of Biomedical Sciences and Medicine (FCBM), University of Algarve, Faro, Portugal
- Centre of Marine Sciences, University of Algarve, Faro, Portugal
| | - Carlos Cardoso
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Porto, Portugal
| | - Jorge Lobo-Arteaga
- Division of Environmental Oceanography, Portuguese Institute for the Sea and Atmosphere, Algés, Portugal
- Marine and Environmental Sciences Centre (MARE), NOVA University of Lisbon, Lisbon, Portugal
| | - Sabrina Sales
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
| | - Diana Juliao
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
| | - Inês Ferreira
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
| | - Paula Chainho
- Marine and Environmental Sciences Centre (MARE), NOVA University of Lisbon, Lisbon, Portugal
| | - Maria Ana Dionísio
- Marine and Environmental Sciences Centre (MARE), NOVA University of Lisbon, Lisbon, Portugal
| | - Maria J. Gaudêncio
- Division of Environmental Oceanography, Portuguese Institute for the Sea and Atmosphere, Algés, Portugal
| | - Cláudia Afonso
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Porto, Portugal
| | - Helena Lourenço
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
| | - M. Leonor Cancela
- Faculty of Biomedical Sciences and Medicine (FCBM), University of Algarve, Faro, Portugal
- Centre of Marine Sciences, University of Algarve, Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
- Centre for BioMedical Research (CBMR), University of Algarve, Faro, Portugal
| | - Narcisa M. Bandarra
- Division of Aquaculture, Upgrading and Bioprospection, Portuguese Institute for the Sea and Atmosphere (IPMA), Algés, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR/CIMAR), University of Porto, Porto, Portugal
| | - Paulo J. Gavaia
- Faculty of Biomedical Sciences and Medicine (FCBM), University of Algarve, Faro, Portugal
- Centre of Marine Sciences, University of Algarve, Faro, Portugal
| |
Collapse
|
24
|
Wu WJ, Xia CL, Ou SJ, Yang Y, Zhou XZ, Ma YF, Hou YL, Wang FZ, Yang QP, Qi Y, Xu CP. Prophylactic Effects of NFκB Essential Modulator–Binding Domain Peptides on Bone Infection: An Experimental Study in a Rabbit Model. J Inflamm Res 2022; 15:2745-2759. [PMID: 35509324 PMCID: PMC9059993 DOI: 10.2147/jir.s346627] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Methods Results Conclusion
Collapse
Affiliation(s)
- Wen-Jiao Wu
- Department of Orthopaedics, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, People’s Republic of China
| | - Chang-Liang Xia
- Department of Orthopaedics, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, People’s Republic of China
| | - Shuan-Ji Ou
- Department of Orthopaedics, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, People’s Republic of China
| | - Yang Yang
- Department of Orthopaedics, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, People’s Republic of China
| | - Xiao-Zhong Zhou
- Department of Orthopaedics, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, People’s Republic of China
| | - Yun-Fei Ma
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Yi-Long Hou
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Fa-Zheng Wang
- Department of Orthopaedics, First People’s Hospital of Kashgar Prefecture, Kashgar, Xinjiang, People’s Republic of China
| | - Qing-Po Yang
- Department of Orthopaedics, First People’s Hospital of Kashgar Prefecture, Kashgar, Xinjiang, People’s Republic of China
| | - Yong Qi
- Department of Orthopaedics, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, People’s Republic of China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Correspondence: Yong Qi; Chang-Peng Xu, Tel +86-20-8916-8085, Email ;
| | - Chang-Peng Xu
- Department of Orthopaedics, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, People’s Republic of China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Correspondence: Yong Qi; Chang-Peng Xu, Tel +86-20-8916-8085, Email ;
| |
Collapse
|
25
|
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
|
26
|
Marino S, Hannemann N, Bishop RT, Zeng F, Carrasco G, Meurisse S, Li B, Sophocleous A, Sparatore A, Baeuerle T, Vukicevic S, Auberval M, Mollat P, Bozec A, Idris AI. Anti-inflammatory, but not osteoprotective, effect of the TRAF6/CD40 inhibitor 6877002 in rodent models of local and systemic osteolysis. Biochem Pharmacol 2021; 195:114869. [PMID: 34896056 DOI: 10.1016/j.bcp.2021.114869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/19/2022]
Abstract
NFκB plays a key role in inflammation and skeletal disorders. Previously, we reported that pharmacological inhibition of NFκB at the level of TRAF6 suppressed RANKL, CD40L and IL1β-induced osteoclastogenesis and attenuated cancer-induced bone disease. TNFα is also known to regulate TRAF6/NFκB signalling, however the anti-inflammatory and osteoprotective effects associated with inhibition of the TNFα/TRAF6/NFκB axis have not been investigated. Here, we show that in vitro and ex vivo exposure to the verified small-molecule inhibitor of TRAF6, 6877002 prevented TNFα-induced NFκB activation, osteoclastogenesis and calvarial osteolysis, but it had no effects on TNFα-induced apoptosis or growth inhibition in osteoblasts. Additionally, 6877002 disrupted T-cells support for osteoclast formation and synoviocyte motility, without affecting the viability of osteoblasts in the presence of T-cells derived factors. Using the collagen-induced arthritis model, we show that oral and intraperitoneal administration of 6877002 in mice reduced joint inflammation and arthritis score. Unexpectedly, no difference in trabecular and cortical bone parameters were detected between vehicle and 6877002 treated mice, indicating lack of osteoprotection by 6877002 in the arthritis model described. Using two independent rodent models of osteolysis, we confirmed that 6877002 had no effect on trabecular and cortical bone loss in both osteoporotic rats or RANKL- treated mice. In contrast, the classic anti-osteolytic alendronate offered complete osteoprotection in RANKL- treated mice. In conclusion, TRAF6 inhibitors may be of value in the management of the inflammatory component of bone disorders, but may not offer protection against local or systemic bone loss, unless combined with anti-resorptive therapy such as bisphosphonates.
Collapse
Affiliation(s)
- Silvia Marino
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK; Bone and Cancer Group, Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XR, UK
| | - Nicole Hannemann
- Department of Internal Medicine, 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ryan T Bishop
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK
| | - Feier Zeng
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK
| | - Giovana Carrasco
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK
| | - Sandrine Meurisse
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Boya Li
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK
| | - Antonia Sophocleous
- Department of Life Sciences, School of Sciences, European University Cyprus, 6 Diogenes Street, 1516 Nicosia, Cyprus
| | - Anna Sparatore
- University of Milano, Department of Pharmaceutical Science, Milan, Italy
| | - Tobias Baeuerle
- Institute of Radiology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum, Erlangen, Bayern, Germany
| | - Slobodan Vukicevic
- Department of Anatomy, Medical School, University of Zagreb, Zagreb, Croatia
| | - Marielle Auberval
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Patrick Mollat
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Aline Bozec
- Department of Internal Medicine, 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aymen I Idris
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK; Bone and Cancer Group, Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XR, UK.
| |
Collapse
|
27
|
Abraham AG, Sun J, Sharma A, Yin MT, Brown JK, Demehri S, Garza J, Shah JG, Palella FJ, Kingsley L, Jamieson BD, Althoff KN, Brown TT. The combined effects of age and HIV on the anatomic distribution of cortical and cancellous bone in the femoral neck among men and women. AIDS 2021; 35:2513-2522. [PMID: 34482349 PMCID: PMC8649032 DOI: 10.1097/qad.0000000000003061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To investigate HIV-related and age-related differences in hip bone structure in men and women. DESIGN Cross sectional study of bone structure and HIV serostatus. METHODS We used Quantitative Computed Tomography (QCT) data from the Multicenter AIDS Cohort Study (MACS) and Women's Interagency HIV Study (WIHS) to examine cortical thickness (CT) and cortical (CBMD), trabecular (TBMD), and integral (IBMD) bone mineral density across anatomic quadrants of the femoral neck in older adult MSM and women with (PWH) and without (PWOH) HIV infection. The percentage difference (%diff) in the means for CT and BMD overall and by quadrant between PWH and PWOH were estimated. RESULTS Among 322 MSM (median age 60 years) with bone measures, distributions were similar between HIV serostatus groups with %diff in the quadrant means ranging from -7 to -1% for CT and from -1 to 4% for BMD, and overall lower hip cortical thickness than expected. In contrast, in 113 women (median age 51 years), PWH had lower CT, IBMD and TBMD consistently across all quadrants, with differences ranging from -10 to -20% for CT, -6 to -11% for IBMD and -3 to -6% for TBMD. Estimates reached statistical significance in superoanterior quadrant for CT and IBMD and inferoposterior for CT. CONCLUSION Among women, PWH appear to have a thinner cortex and less dense integral bone compared with PWOH, particularly in the superior quadrants whereas MSM overall had a thinner than expected hip cortex.
Collapse
Affiliation(s)
- Alison G Abraham
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Denver, Colorado
- Department of Epidemiology, The Johns Hopkins School of Public Health, Baltimore, Maryland
| | - Jing Sun
- Department of Epidemiology, The Johns Hopkins School of Public Health, Baltimore, Maryland
| | - Anjali Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Michael T Yin
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Shadpour Demehri
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joshua Garza
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jayesh G Shah
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, New York, USA
| | - Frank J Palella
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Lawrence Kingsley
- Infectious Diseases and Microbiology Department, University of Pittsburgh, Pennsylvania
| | - Beth D Jamieson
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Keri N Althoff
- Department of Epidemiology, The Johns Hopkins School of Public Health, Baltimore, Maryland
| | - Todd T Brown
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
28
|
Yang T, Sun K, Wang C, Swarnkar G, Quan S, Kress D, Xiao J, Alippe Y, Zheng H, Brophy RH, Hao D, McAlinden A, Abu-Amer Y, Shen J, Mbalaviele G. Gasdermin D deficiency attenuates arthritis induced by traumatic injury but not autoantibody-assembled immune complexes. Arthritis Res Ther 2021; 23:286. [PMID: 34784954 PMCID: PMC8594229 DOI: 10.1186/s13075-021-02668-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/31/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gasdermin D (GSDMD) is cleaved by several proteases including by caspase-1, a component of intracellular protein complexes called inflammasomes. Caspase-1 also converts pro-interleukin-1β (pro-IL-1β) and pro-IL-18 into bioactive IL-1β and IL-18, respectively. GSDMD amino-terminal fragments form plasma membrane pores, which mediate the secretion of IL-1β and IL-18 and cause the inflammatory form of cell death pyroptosis. Here, we tested the hypothesis that GSDMD contributes to joint degeneration in the K/BxN serum transfer-induced arthritis (STIA) model in which autoantibodies against glucose-6-phosphate isomerase promote the formation of pathogenic immune complexes on the surface of myeloid cells, which highly express the inflammasomes. The unexpected outcomes with the STIA model prompted us to determine the role of GSDMD in the post-traumatic osteoarthritis (PTOA) model caused by meniscus ligamentous injury (MLI) based on the hypothesis that this pore-forming protein is activated by signals released from damaged joint tissues. METHODS Gsdmd +/+ and Gsdmd-/- mice were injected with K/BxN mouse serum or subjected to MLI to cause STIA or PTOA, respectively. Paw and ankle swelling and DXA scanning were used to assess the outcomes in the STIA model whereas histopathology and micro-computed tomography (μCT) were utilized to monitor joints in the PTOA model. Murine and human joint tissues were also examined for GSDMD, IL-1β, and IL-18 expression by qPCR, immunohistochemistry, or immunoblotting. RESULTS GSDMD levels were higher in serum-inoculated paws compared to PBS-injected paws. Unexpectedly, ablation of GSDMD failed to reduce joint swelling and osteolysis, suggesting that GSDMD was dispensable for the pathogenesis of STIA. GSDMD levels were also higher in MLI compared to sham-operated joints. Importantly, ablation of GSDMD attenuated MLI-associated cartilage degradation (p = 0.0097), synovitis (p = 0.014), subchondral bone sclerosis (p = 0.0006), and subchondral bone plate thickness (p = 0.0174) based on histopathological and μCT analyses. CONCLUSION GSDMD plays a key role in the pathogenesis of PTOA, but not STIA, suggesting that its actions in experimental arthropathy are tissue context-specific.
Collapse
Affiliation(s)
- Tong Yang
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Kai Sun
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Chun Wang
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Gaurav Swarnkar
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Songtao Quan
- Luoyang Orthopedic - Traumatological Hospital of Henan Province, Luoyang, Henan, China
| | - Dustin Kress
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Jianqiu Xiao
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Yael Alippe
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Hongjun Zheng
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert H Brophy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Dingjun Hao
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospital for Children, St. Louis, MO, USA
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospital for Children, St. Louis, MO, USA
| | - Jie Shen
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA.
| |
Collapse
|
29
|
Gao X, Wu Q, Zhang X, Tian J, Liang D, Min Y, Lu J, Zhang X, Cui L, Xu B, Liu Y. Salvianolate Ameliorates Osteopenia and Improves Bone Quality in Prednisone-Treated Rheumatoid Arthritis Rats by Regulating RANKL/RANK/OPG Signaling. Front Pharmacol 2021; 12:710169. [PMID: 34552485 PMCID: PMC8450458 DOI: 10.3389/fphar.2021.710169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/23/2021] [Indexed: 01/01/2023] Open
Abstract
Rheumatoid arthritis (RA) is closely associated with periarticular osteopenia and leads to a high risk of generalized osteoporosis. Although glucocorticoid (GC) treatment ameliorates joint degradation and manages inflammation in RA, GC application may induce further bone quality deterioration in RA patients. Current treatments for RA lack relevant strategies for the prevention and treatment of osteopenia in RA. In this study, we aimed to investigate whether salvianolate treatment ameliorated osteopenia in prednisone-treated RA rats. Lewis rats with collagen-induced arthritis (CIA) were administered prednisone (PDN) or PDN plus salvianolate (PDN+Sal) treatment for 90 days. The effects of Sal were investigated in PDN-treated CIA rats. To further evaluate the effects of Sal under inflammatory conditions, we investigated the effects of Sal treatment on the TNF-α-induced inflammatory response in MC3T3-E1 osteoblasts. Bone histomorphometry, bone mineral density (BMD), bone biomechanical properties, micro-computed tomography (micro-CT), immunohistochemistry, RT-PCR and western blot analyses were performed to evaluate the effects of Sal. The results demonstrated that RA induced bone loss and bone quality deterioration, with high bone turnover in CIA rats. PDN+Sal treatment significantly increased BMD and trabecular/cortical bone mass, suppressed inflammation, and improved bone biomechanical properties compared to CIA control and PDN treatment. PDN+Sal treatment significantly suppressed bone resorption and the RANKL and RANKL/OPG ratios compared to PDN. PDN+Sal and PDN treatment significantly inhibited TNF-α by 82 and 83%, respectively, and both suppressed inflammation in CIA rats. However, there was no significant difference between PDN+Sal and PDN treatment alone in regard to bone formation parameters or the management of inflammation and arthropathy. Sal significantly increased Osterix, OPN, and Col1a1 while decreasing RANKL, TRAF6, and TRAIL gene in TNF-α-induced MC3T3-E1 osteoblasts. Sal significantly increased Osterix, OPN and RUNX2 while decreasing NF-κB, TRAF6 and IL-1β protein in TNF-α-induced MC3T3-E1 osteoblasts. The results suggested that salvianolate treatment ameliorated osteopenia and improved bone quality in prednisone-treated RA rats, and the potential mechanism may be related to the regulation of the RANKL/RANK/OPG signaling pathway, TRAIL-TRAF6-NFκB signal axis, and downregulation of inflammatory cytokines. Salvianolate could be used as a promising supplemental therapeutic strategy to ameliorate osteopenia and improve bone quality in GC-treated RA patients.
Collapse
Affiliation(s)
- Xiang Gao
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical University, Zhanjiang, China.,Stem Cell Research and Cellular Therapy Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qingyun Wu
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical University, Zhanjiang, China.,Department of Pharmacy, Yangjiang People's Hospital, Yangjiang, China
| | - Xinle Zhang
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical University, Zhanjiang, China
| | - Jia Tian
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical University, Zhanjiang, China
| | - Dahong Liang
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical University, Zhanjiang, China
| | - Yalin Min
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical University, Zhanjiang, China
| | - Jiaqi Lu
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical University, Zhanjiang, China
| | - Xuemei Zhang
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical University, Zhanjiang, China
| | - Liao Cui
- Stem Cell Research and Cellular Therapy Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Bilian Xu
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical University, Zhanjiang, China
| | - Yanzhi Liu
- Department of Pharmacology, Guangdong Key Laboratory for Research and Development of Natural Drug, Guangdong Medical University, Zhanjiang, China.,Clinic Research Institute of Zhanjiang, Affiliated Central People's Hospital of Zhanjiang of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
30
|
Chellaiah MA. L-Plastin Phosphorylation: Possible Regulation by a TNFR1 Signaling Cascade in Osteoclasts. Cells 2021; 10:2432. [PMID: 34572081 PMCID: PMC8464874 DOI: 10.3390/cells10092432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/25/2021] [Accepted: 09/09/2021] [Indexed: 12/30/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) signaling regulates phosphorylation of L-plastin, which is involved in forming the nascent sealing zone, a precursor zone for the matured sealing ring. This study aimed to illustrate the molecular mechanisms of L-plastin phosphorylation and the subsequent formation of the nascent sealing zone in osteoclasts treated with TNF-α. Here, we report that anti-TNF-receptor 1, inhibitors of signaling proteins (Src, PI3-K, Rho, and Rho-kinase), and siRNA of TRAF-6 attenuated the phosphorylation of LPL and filamentous actin content significantly in the presence of TNF-α. An inhibitor of integrin αvβ3, PKC, or PKA did not inhibit TNF-α-induced L-plastin phosphorylation. Inhibitors of Src and PI3-K and not Rho or Rho-kinase reduced tyrosine phosphorylation of TRAF-6, suggesting that Src and PI3-K regulate TRAF-6 phosphorylation, and Rho and Rho-kinase are downstream of TRAF-6 regulation. Osteoclasts expressing constitutively active or kinase-defective Src proteins were used to determine the role of Src on L-plastin phosphorylation; similarly, the effect of Rho was confirmed by transducing TAT-fused constitutively active (V14) or dominant-negative (N19) Rho proteins into osteoclasts. Pull-down analysis with glutathione S-transferase-fused SH2 and SH3 domains of Src and PI3-K demonstrated coprecipitation of L-plastin and TRAF-6 with the SH3 and SH2 domains of the PI3-K and Src proteins. However, the actual order of the interaction of proteins requires further elucidation; a comprehensive screening should corroborate the initial findings of protein interactions via the SH2/SH3 domains. Ultimately, inhibition of the interaction of proteins with SH2/SH3 could reduce L-plastin phosphorylation and affect NSZ formation and bone resorption in conditions that display osteoclast activation and bone loss.
Collapse
Affiliation(s)
- Meenakshi A Chellaiah
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
31
|
Charoenngam N, Ponvilawan B, Thongpiya J, Yingchoncharoen P, Ungprasert P. Psoriatic Arthritis and Risk of Vertebral Fracture: A Systematic Review and Meta-analysis. Curr Rheumatol Rev 2021; 18:64-71. [PMID: 34496734 DOI: 10.2174/1573397117666210908094349] [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: 10/29/2020] [Revised: 05/18/2021] [Accepted: 06/07/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study was conducted in order to determine the association between psoriatic arthritis and risk of vertebral fracture by pooling the evidence from previous studies. METHODS Potentially eligible studies were identified from MEDLINE and EMBASE database from inception to March 2020 using search strategy that comprised of terms for "Psoriatic Arthritis" and "Vertebral Fracture". Studies were eligible for the meta-analysis if they were cohort studies that included psoriatic arthritis and individuals without psoriasis and followed them for incident vertebral fracture. Studies were also required to report standardized incidence ration, hazard risk ratio or relative risk with related 95% confidence intervals (CI) comparing the incidence of vertebral fracture between the two cohorts. The retrieved point estimates with standard errors from each study were pooled into the final result by the random-effect model, generic inverse variance method. RESULTS A total of 26,090 articles were identified. After two rounds of independent review by three investigators, we included five cohort studies that met the eligibility criteria in the meta-analysis. PsA is significantly associated with VF the pooled odds ratio of 2.09 (95% CI, 1.11 - 3.96; I2 70%). The funnel plot was fairly asymmetric, thus, the publication bias in favor of studies may present. CONCLUSIONS This systematic review and meta-analysis indicates that psoriatic arthritis patients have a significantly elevated risk of developing vertebral fracture.
Collapse
Affiliation(s)
- Nipith Charoenngam
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Thailand
| | - Ben Ponvilawan
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Thailand
| | - Jerapas Thongpiya
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Thailand
| | - Pitchaporn Yingchoncharoen
- Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok. Thailand
| | - Patompong Ungprasert
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic Foundation, Cleveland, OH. United States
| |
Collapse
|
32
|
Schreuder WH, van der Wal JE, de Lange J, van den Berg H. Multiple versus solitary giant cell lesions of the jaw: Similar or distinct entities? Bone 2021; 149:115935. [PMID: 33771761 DOI: 10.1016/j.bone.2021.115935] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/27/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023]
Abstract
The majority of giant cell lesions of the jaw present as a solitary focus of disease in bones of the maxillofacial skeleton. Less frequently they occur as multifocal lesions. This raises the clinical dilemma if these should be considered distinct entities and therefore each need a specific therapeutic approach. Solitary giant cell lesions of the jaw present with a great diversity of symptoms. Recent molecular analysis revealed that these are associated with somatic gain-of-function mutations in KRAS, FGFR1 or TRPV4 in a large component of the mononuclear stromal cells which all act on the RAS/MAPK pathway. For multifocal lesions, a small group of neoplastic multifocal giant cell lesions of the jaw remain after ruling out hyperparathyroidism. Strikingly, most of these patients are diagnosed with jaw lesions before the age of 20 years, thus before the completion of dental and jaw development. These multifocal lesions are often accompanied by a diagnosis or strong clinical suspicion of a syndrome. Many of the frequently reported syndromes belong to the so-called RASopathies, with germline or mosaic mutations leading to downstream upregulation of the RAS/MAPK pathway. The other frequently reported syndrome is cherubism, with gain-of-function mutations in the SH3BP2 gene leading through assumed and unknown signaling to an autoinflammatory bone disorder with hyperactive osteoclasts and defective osteoblastogenesis. Based on this extensive literature review, a RAS/MAPK pathway activation is hypothesized in all giant cell lesions of the jaw. The different interaction between and contribution of deregulated signaling in individual cell lineages and crosstalk with other pathways among the different germline- and non-germline-based alterations causing giant cell lesions of the jaw can be explanatory for the characteristic clinical features. As such, this might also aid in the understanding of the age-dependent symptomatology of syndrome associated giant cell lesions of the jaw; hopefully guiding ideal timing when installing treatment strategies in the future.
Collapse
Affiliation(s)
- Willem H Schreuder
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC and Academic Center for Dentistry Amsterdam, University of Amsterdam, Amsterdam, the Netherlands; Department of Head and Neck Surgery and Oncology, Antoni van Leeuwenhoek / Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Jacqueline E van der Wal
- Department of Pathology, Antoni van Leeuwenhoek / Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jan de Lange
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC and Academic Center for Dentistry Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Henk van den Berg
- Department of Pediatrics / Oncology, Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Amsterdam, the Netherlands
| |
Collapse
|
33
|
Yan T, Xie Y, He H, Fan W, Huang F. Role of nitric oxide in orthodontic tooth movement (Review). Int J Mol Med 2021; 48:168. [PMID: 34278439 PMCID: PMC8285047 DOI: 10.3892/ijmm.2021.5001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Nitric oxide (NO) is an ubiquitous signaling molecule that mediates numerous cellular processes associated with cardiovascular, nervous and immune systems. NO also plays an essential role in bone homeostasis regulation. The present review article summarized the effects of NO on bone metabolism during orthodontic tooth movement in order to provide insight into the regulatory role of NO in orthodontic tooth movement. Orthodontic tooth movement is a process in which the periodontal tissue and alveolar bone are reconstructed due to the effect of orthodontic forces. Accumulating evidence has indicated that NO and its downstream signaling molecule, cyclic guanosine monophosphate (cGMP), mediate the mechanical signals during orthodontic-related bone remodeling, and exert complex effects on osteogenesis and osteoclastogenesis. NO has a regulatory effect on the cellular activities and functional states of osteoclasts, osteocytes and periodontal ligament fibroblasts involved in orthodontic tooth movement. Variations of NO synthase (NOS) expression levels and NO production in periodontal tissues or gingival crevicular fluid (GCF) have been found on the tension and compression sides during tooth movement in both orthodontic animal models and patients. Furthermore, NO precursor and NOS inhibitor administration increased and reduced the tooth movement in animal models, respectively. Further research is required in order to further elucidate the underlying mechanisms and the clinical application prospect of NO in orthodontic tooth movement.
Collapse
Affiliation(s)
- Tong Yan
- Department of Pediatric Dentistry, Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Yongjian Xie
- Department of Orthodontic Dentistry, Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wenguo Fan
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Fang Huang
- Department of Pediatric Dentistry, Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| |
Collapse
|
34
|
Salivary Pro-Inflammatory Markers and Smoking Status Influences the Treatment Effectiveness of Periodontal Disease Patients with Hypertension. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18147364. [PMID: 34299815 PMCID: PMC8305443 DOI: 10.3390/ijerph18147364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/24/2021] [Accepted: 07/06/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Hypertension and periodontal diseases share several risk factors. Inflammation biomarkers in saliva are related to hypertension and periodontal disease. The aim of this study was to explore the role of the salivary inflammatory biomarkers in the treatment effectiveness of patients with hypertension and periodontal disease. METHODS This observational study enrolled 160 subjects diagnosed with periodontitis, 40 of which had a history of hypertension. All subjects had completed scaling and root planning therapeutic procedures within four weeks. The clinical periodontal parameters (i.e., bleeding on probing, plaque control record (PCR), and probing depth (PD)) were evaluated before and after the treatment. Pro-inflammatory markers were determined using a commercial kit. RESULTS The recovery rate (PD 4-9 mm) in non-hypertensive subjects was significantly higher than in hypertensive subjects (60.47% vs. 52.60%, respectively; p = 0.04). All clinical parameters, excluding PCR, positively correlated with salivary IL-1β at baseline and after completing treatment. Our results showed that increased salivary IL-1β levels were positively associated with decreased PCR (β = -27.65 and p = 0.05) and PD recovery rate (β = -17.05 and p = 0.02) in hypertensive subjects. CONCLUSIONS The present study sheds important light on the clinical use of salivary pro-inflammatory cytokines as valuable biomarkers for predicting the treatment effectiveness of patients suffering from hypertension and periodontitis.
Collapse
|
35
|
Patil KC, Soekmadji C. Extracellular Vesicle-Mediated Bone Remodeling and Bone Metastasis: Implications in Prostate Cancer. Subcell Biochem 2021; 97:297-361. [PMID: 33779922 DOI: 10.1007/978-3-030-67171-6_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone metastasis is the tendency of certain primary tumors to spawn and dictate secondary neoplasia in the bone. The process of bone metastasis is regulated by the dynamic crosstalk between metastatic cancer cells, cellular components of the bone marrow microenvironment (osteoblasts, osteoclasts, and osteocytes), and the bone matrix. The feed-forward loop mechanisms governs the co-option of homeostatic bone remodeling by cancer cells in bone. Recent developments have highlighted the discovery of extracellular vesicles (EVs) and their diverse roles in distant outgrowths. Several studies have implicated EV-mediated interactions between cancer cells and the bone microenvironment in synergistically promoting pathological skeletal metabolism in the metastatic site. Nevertheless, the potential role that EVs serve in arbitrating intricate sequences of coordinated events within the bone microenvironment remains an emerging field. In this chapter, we review the role of cellular participants and molecular mechanisms in regulating normal bone physiology and explore the progress of current research into bone-derived EVs in directly triggering and coordinating the processes of physiological bone remodeling. In view of the emerging role of EVs in interorgan crosstalk, this review also highlights the multiple systemic pathophysiological processes orchestrated by the EVs to direct organotropism in bone in prostate cancer. Given the deleterious consequences of bone metastasis and its clinical importance, in-depth knowledge of the multifarious role of EVs in distant organ metastasis is expected to open new possibilities for prognostic evaluation and therapeutic intervention for advanced bone metastatic prostate cancer.
Collapse
Affiliation(s)
- Kalyani C Patil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia. .,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
36
|
AlQranei MS, Senbanjo LT, Aljohani H, Hamza T, Chellaiah MA. Lipopolysaccharide- TLR-4 Axis regulates Osteoclastogenesis independent of RANKL/RANK signaling. BMC Immunol 2021; 22:23. [PMID: 33765924 PMCID: PMC7995782 DOI: 10.1186/s12865-021-00409-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 03/01/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Lipopolysaccharide (LPS) is an endotoxin and a vital component of gram-negative bacteria's outer membrane. During gram-negative bacterial sepsis, LPS regulates osteoclast differentiation and activity, in addition to increasing inflammation. This study aimed to investigate how LPS regulates osteoclast differentiation of RAW 264.7 cells in vitro. RESULTS Herein, we revealed that RAW cells failed to differentiate into mature osteoclasts in vitro in the presence of LPS. However, differentiation occurred in cells primed with receptor activator of nuclear factor-kappa-Β ligand (RANKL) for 24 h and then treated with LPS for 48 h (henceforth, denoted as LPS-treated cells). In cells treated with either RANKL or LPS, an increase in membrane levels of toll-like receptor 4 (TLR4) receptor was observed. Mechanistically, an inhibitor of TLR4 (TAK-242) reduced the number of osteoclasts as well as the secretion of tumor necrosis factor (TNF)-α in LPS-treated cells. RANKL-induced RAW cells secreted a very basal level TNF-α. TAK-242 did not affect RANKL-induced osteoclastogenesis. Increased osteoclast differentiation in LPS-treated osteoclasts was not associated with the RANKL/RANK/OPG axis but connected with the LPS/TLR4/TNF-α tumor necrosis factor receptor (TNFR)-2 axis. We postulate that this is because TAK-242 and a TNF-α antibody suppress osteoclast differentiation. Furthermore, an antibody against TNF-α reduced membrane levels of TNFR-2. Secreted TNF-α appears to function as an autocrine/ paracrine factor in the induction of osteoclastogenesis independent of RANKL. CONCLUSION TNF-α secreted via LPS/TLR4 signaling regulates osteoclastogenesis in macrophages primed with RANKL and then treated with LPS. Our findings suggest that TLR4/TNF-α might be a potential target to suppress bone loss associated with inflammatory bone diseases, including periodontitis, rheumatoid arthritis, and osteoporosis.
Collapse
Affiliation(s)
- Mohammed S AlQranei
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, 650 W Baltimore Street, Baltimore, MD, 21201, USA
- Preventive Dental Sciences Department, School of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Linda T Senbanjo
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, 650 W Baltimore Street, Baltimore, MD, 21201, USA
| | - Hanan Aljohani
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, 650 W Baltimore Street, Baltimore, MD, 21201, USA
- Department of Oral Medicine and Diagnostics Sciences, King Saud University, School of Dentistry, Riyadh, Kingdom of Saudi Arabia
| | - Therwa Hamza
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, 650 W Baltimore Street, Baltimore, MD, 21201, USA
| | - Meenakshi A Chellaiah
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, 650 W Baltimore Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
37
|
Computational modeling reveals a key role for polarized myeloid cells in controlling osteoclast activity during bone injury repair. Sci Rep 2021; 11:6055. [PMID: 33723343 PMCID: PMC7961065 DOI: 10.1038/s41598-021-84888-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/11/2021] [Indexed: 01/12/2023] Open
Abstract
Bone-forming osteoblasts and -resorbing osteoclasts control bone injury repair, and myeloid-derived cells such as monocytes and macrophages are known to influence their behavior. However, precisely how these multiple cell types coordinate and regulate each other over time within the bone marrow to restore bone is difficult to dissect using biological approaches. Conversely, mathematical modeling lends itself well to this challenge. Therefore, we generated an ordinary differential equation (ODE) model powered by experimental data (osteoblast, osteoclast, bone volume, pro- and anti-inflammatory myeloid cells) obtained from intra-tibially injured mice. Initial ODE results using only osteoblast/osteoclast populations demonstrated that bone homeostasis could not be recovered after injury, but this issue was resolved upon integration of pro- and anti-inflammatory myeloid population dynamics. Surprisingly, the ODE revealed temporal disconnects between the peak of total bone mineralization/resorption, and osteoblast/osteoclast numbers. Specifically, the model indicated that osteoclast activity must vary greatly (> 17-fold) to return the bone volume to baseline after injury and suggest that osteoblast/osteoclast number alone is insufficient to predict bone the trajectory of bone repair. Importantly, the values of osteoclast activity fall within those published previously. These data underscore the value of mathematical modeling approaches to understand and reveal new insights into complex biological processes.
Collapse
|
38
|
Ansalone C, Cole J, Chilaka S, Sunzini F, Sood S, Robertson J, Siebert S, McInnes IB, Goodyear CS. TNF is a homoeostatic regulator of distinct epigenetically primed human osteoclast precursors. Ann Rheum Dis 2021; 80:748-757. [PMID: 33692019 PMCID: PMC8142443 DOI: 10.1136/annrheumdis-2020-219262] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/31/2020] [Accepted: 01/25/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Circulating myeloid precursors are responsible for post-natal osteoclast (OC) differentiation and skeletal health, although the exact human precursors have not been defined. Enhanced osteoclastogenesis contributes to joint destruction in rheumatoid arthritis (RA) and tumour necrosis factor (TNF) is a well-known pro-osteoclastogenic factor. Herein, we investigated the interplay between receptor activator of nuclear factor kappa-Β ligand (RANK-L), indispensable for fusion of myeloid precursors and the normal development of OCs, and TNF in directing the differentiation of diverse pre-OC populations derived from human peripheral blood. METHODS Flow cytometric cell sorting and analysis was used to assess the potential of myeloid populations to differentiate into OCs. Transcriptomic, epigenetic analysis, receptor expression and inhibitor experiments were used to unravel RANK-L and TNF signalling hierarchy. RESULTS TNF can act as a critical homoeostatic regulator of CD14+ monocyte (MO) differentiation into OCs by inhibiting osteoclastogenesis to favour macrophage development. In contrast, a distinct previously unidentified CD14-CD16-CD11c+ myeloid pre-OC population was exempt from this negative regulation. In healthy CD14+ MOs, TNF drove epigenetic modification of the RANK promoter via a TNFR1-IKKβ-dependent pathway and halted osteoclastogenesis. In a subset of patients with RA, CD14+ MOs exhibited an altered epigenetic state that resulted in dysregulated TNF-mediated OC homoeostasis. CONCLUSIONS These findings fundamentally re-define the relationship between RANK-L and TNF. Moreover, they have identified a novel pool of human circulating non-MO OC precursors that unlike MOs are epigenetically preconditioned to ignore TNF-mediated signalling. In RA, this epigenetic preconditioning occurs in the MO compartment providing a pathological consequence of failure of this pathway.
Collapse
Affiliation(s)
- Cecilia Ansalone
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - John Cole
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Sabarinadh Chilaka
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Flavia Sunzini
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Shatakshi Sood
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Jamie Robertson
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Stefan Siebert
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Carl S Goodyear
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
39
|
Jia X, Zhu H, Li G, Lan M, Li X, Huang M, Xu W, Wu S. Anti-osteoporotic effects of alisol C 23-acetate via osteoclastogenesis inhibition. Biomed Pharmacother 2021; 137:111321. [PMID: 33524783 DOI: 10.1016/j.biopha.2021.111321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/28/2020] [Accepted: 01/21/2021] [Indexed: 12/21/2022] Open
Abstract
Alismatis rhizoma (AR) is the dried rhizome of Alisma orientale (Sam.) Juz. (Alismataceae). This traditional Chinese formula is diuretic, hypoglycemic, and hypolipidemic. Alisol C 23-acetate (AC23A) from AR is anti-inflammatory and ameliorates certain metabolic diseases. However, the mechanism by which AC23A mitigates osteoporosis is unknown. The present study investigated the anti-osteoporotic effects of AC23A in vivo and in vitro. In an ovariectomized (OVX) rat model, AC23A ameliorated OVX-induced organ coefficients and trabecular bone loss. In OVX rats, AC23A treatment lowered serum TRAP5b, CTK, β-CTX, TNF-α, IL-6, and IL-1β, raised serum E2, and did not significantly change serum OCN or BALP. AC23A inhibited osteoclast formation in a rat co-culture system without affecting osteoblast activity. RANK (receptor activator of nuclear factor kappaB) signaling channels are vital osteoclastogenesis transcription elements. AC23A inhibited RANK ligand (RANKL)-induced TRAP, c-Fos, MMP9, NFATc1, and CTK expression and JNK phosphorylation. Therefore, AC23A is anti-osteoclastogenic in vitro and in vivo by inhibiting RANKL-induced osteoclast differentiation and function. Moreover, AC23A could help prevent or limit osteoclast-mediated bone diseases by inhibiting osteoclastogenesis.
Collapse
Affiliation(s)
- Xiaokang Jia
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Huaichang Zhu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Gaopan Li
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Mengliu Lan
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Xiaoyan Li
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Mingqing Huang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Wen Xu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Shuisheng Wu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| |
Collapse
|
40
|
Ng JS, Chin KY. Potential mechanisms linking psychological stress to bone health. Int J Med Sci 2021; 18:604-614. [PMID: 33437195 PMCID: PMC7797546 DOI: 10.7150/ijms.50680] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic psychological stress affects many body systems, including the skeleton, through various mechanisms. This review aims to provide an overview of the factors mediating the relationship between psychological stress and bone health. These factors can be divided into physiological and behavioural changes induced by psychological stress. The physiological factors involve endocrinological changes, such as increased glucocorticoids, prolactin, leptin and parathyroid hormone levels and reduced gonadal hormones. Low-grade inflammation and hyperactivation of the sympathetic nervous system during psychological stress are also physiological changes detrimental to bone health. The behavioural changes during mental stress, such as altered dietary pattern, cigarette smoking, alcoholism and physical inactivity, also threaten the skeletal system. Psychological stress may be partly responsible for epigenetic regulation of skeletal development. It may also mediate the relationship between socioeconomic status and bone health. However, more direct evidence is required to prove these hypotheses. In conclusion, chronic psychological stress should be recognised as a risk factor of osteoporosis and stress-coping methods should be incorporated as part of the comprehensive osteoporosis-preventing strategy.
Collapse
Affiliation(s)
- Jia-Sheng Ng
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Malaysia
| |
Collapse
|
41
|
Freire MS, Oliveira NG, Lima SMF, Porto WF, Martins DCM, Silva ON, Chaves SB, Sousa MV, Ricart CAO, Castro MS, Fontes W, Franco OL, Rezende TMB. IL-4 absence triggers distinct pathways in apical periodontitis development. J Proteomics 2020; 233:104080. [PMID: 33338687 DOI: 10.1016/j.jprot.2020.104080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/17/2020] [Accepted: 12/12/2020] [Indexed: 12/18/2022]
Abstract
Dental pulp is a specialized tissue able to respond to infectious processes. Nevertheless, infection progress and root canal colonization trigger an immune-inflammatory response in tooth-surrounding tissues, leading to apical periodontitis and bone tissue destruction, further contributing to tooth loss. In order to shed some light on the effects of IL-4 on periradicular pathology development modulation, microtomographic, histological and proteomic analyses were performed using 60 mice, 30 wild type and 30 IL-4-/-. For that, 5 animals were used for microtomographic and histological analysis, and another 5 for proteomic analysis for 0, 7 and 21 days with/without pulp exposure. The periapical lesions were established in WT and IL-4-/- mice without statistical differences in their volume, and the value of p < 0.05 was adopted as significant in microtomographic and histological analyses. Regarding histological analysis, IL-4-/- mice show aggravation of pulp inflammation compared to WT. By using proteomic analysis, we have identified 32 proteins with increased abundance and 218 proteins with decreased abundance in WT animals after 21 days of pulp exposure, compared to IL-4-/- animals. However, IL-4-/- mice demonstrated faster development of apical periodontitis. These animals developed a compensatory mechanism to overcome IL-4 absence, putatively based on the identification of upregulated proteins related to immune system signaling pathways. Significance: IL-4 might play a protective role in diseases involving bone destruction and its activity may contribute to host protection, mainly due to its antiosteoclastogenic action.
Collapse
Affiliation(s)
- Mirna S Freire
- Programa de Pós-Graduação em Biotecnologia e Biodiversidade, Universidade de Brasília, Brasília, DF, Brazil; Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; Curso de Odontologia, Centro Universitário do Planalto Central Apparecido dos Santos, UNICEPLAC, Brasília, DF, Brazil
| | - Nelson G Oliveira
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil
| | - Stella M F Lima
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; Curso de Odontologia, Universidade Católica de Brasília, UCB, Brasília, DF, Brazil
| | - William F Porto
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; Porto Reports, Brasília, DF, Brazil
| | - Danilo C M Martins
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Ciências da Saúde, Universidade de Brasília, UnB, Brasília, DF, Brazil
| | - Osmar N Silva
- Programa de Pós-graduacao em Ciências Farmacêuticas. Centro Universitário de Anápolis - UniEVANGELICA, Anápolis, GO, Brazil
| | - Sacha B Chaves
- Departamento de nanotecnologia, Universidade de Brasília, Brazil
| | - Marcelo V Sousa
- Laboratório de Bioquímica e Química de Proteínas, Departamento de Biologia Celular, Universidade de Brasília, Brazil
| | - Carlos A O Ricart
- Laboratório de Bioquímica e Química de Proteínas, Departamento de Biologia Celular, Universidade de Brasília, Brazil
| | - Mariana S Castro
- Laboratório de Bioquímica e Química de Proteínas, Departamento de Biologia Celular, Universidade de Brasília, Brazil
| | - Wagner Fontes
- Laboratório de Bioquímica e Química de Proteínas, Departamento de Biologia Celular, Universidade de Brasília, Brazil
| | - Octavio L Franco
- Programa de Pós-Graduação em Biotecnologia e Biodiversidade, Universidade de Brasília, Brasília, DF, Brazil; Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Ciências da Saúde, Universidade de Brasília, UnB, Brasília, DF, Brazil; Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília, Brasília, DF, Brazil.
| | - Taia M B Rezende
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; Curso de Odontologia, Universidade Católica de Brasília, UCB, Brasília, DF, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Ciências da Saúde, Universidade de Brasília, UnB, Brasília, DF, Brazil.
| |
Collapse
|
42
|
Multitasking by the OC Lineage during Bone Infection: Bone Resorption, Immune Modulation, and Microbial Niche. Cells 2020; 9:cells9102157. [PMID: 32987689 PMCID: PMC7598711 DOI: 10.3390/cells9102157] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 01/18/2023] Open
Abstract
Bone infections, also known as infectious osteomyelitis, are accompanied by significant inflammation, osteolysis, and necrosis. Osteoclasts (OCs) are the bone-resorbing cells that work in concert with osteoblasts and osteocytes to properly maintain skeletal health and are well known to respond to inflammation by increasing their resorptive activity. OCs have typically been viewed merely as effectors of pathologic bone resorption, but recent evidence suggests they may play an active role in the progression of infections through direct effects on pathogens and via the immune system. This review discusses the host- and pathogen-derived factors involved in the in generation of OCs during infection, the crosstalk between OCs and immune cells, and the role of OC lineage cells in the growth and survival of pathogens, and highlights unanswered questions in the field.
Collapse
|
43
|
Behl T, Chadha S, Sachdeva M, Kumar A, Hafeez A, Mehta V, Bungau S. Ubiquitination in rheumatoid arthritis. Life Sci 2020; 261:118459. [PMID: 32961230 DOI: 10.1016/j.lfs.2020.118459] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 12/23/2022]
Abstract
Rheumatoid arthritis is a chronic, inflammatory joint disease leading to inflammation of synovial membrane that lines the joints. This inflammation further progresses and results in destruction of joints and surrounding cartilages. The underlying factors can be oxidative stress, pro-inflammatory mediators, imbalance and attenuation between various enzymes and proteins (like nuclear factor erythroid 2 related factor 2/Nrf2 and ubiquitin). Protein degradation pathways comprises of lysosomal, proteasomal pathway, and autophagosome (that are carried out in mammalian cells) are regulated through ubiquitin. Ubiquitin proteasomal system is dominating pathway for carrying out non-lysosomal proteolysis of intracellularly proteins. Fundamental processes including cell cycle progression, process of division, apoptosis, modulation of immune responses and cell trafficking are regulated by process of ubiquitination. Ubiquitin proteasomal pathway (UPP) includes ubiquitin moieties which are covalently attached to proteins and guides them proteasome for degradation. Misfolded, oxidized and damaged proteins which are responsible for critical processes, are major targets of degradation process. Any alteration in this system leads to dysregulated cellular homeostasis; progressively leading to numerous diseases including rheumatoid arthritis. Factors including TAK1, TRAF6 undergo are required for the progression of disease and thus contributes towards pathology of inflammatory disorders such as rheumatoid arthritis. This review will include all linked aspects which contribute its major role in rheumatoid arthritis.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Swati Chadha
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Monika Sachdeva
- Fatima College of Health Sciences, Al Ain, United Arab Emirates
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Mirzapur Pole, Uttar Pradesh, India
| | - Vineet Mehta
- Department of Pharmacology, Government College of Pharmacy, Rohru, Ditt. Shimla, Himachal Pradesh, India
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
44
|
Pham HT, Kram V, Dar QA, Komori T, Ji Y, Mohassel P, Rooney J, Li L, Kilts TM, Bonnemann C, Lamande S, Young MF. Collagen VIα2 chain deficiency causes trabecular bone loss by potentially promoting osteoclast differentiation through enhanced TNFα signaling. Sci Rep 2020; 10:13749. [PMID: 32792616 PMCID: PMC7426410 DOI: 10.1038/s41598-020-70730-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
Type VI collagen is well known for its role in muscular disorders, however its function in bone is still not well understood. To examine its role in bone we analyzed femoral and vertebral bone mass by micro-computed tomography analysis, which showed lower bone volume/total volume and trabecular number in Col6α2-KO mice compared with WT. Dynamic histomorphometry showed no differences in trabecular bone formation between WT and Col6α2-KO mice based on the mineral appositional rate, bone formation rate, and mineralizing perimeter. Femoral sections were assessed for the abundance of Tartrate Resistant Acid Phosphatase-positive osteoclasts, which revealed that mutant mice had more osteoclasts compared with WT mice, indicating that the primary effect of Col6a2 deficiency is on osteoclastogenesis. When bone marrow stromal cells (BMSCs) from WT and Col6α2-KO mice were treated with rmTNFα protein, the Col6α2-KO cells expressed higher levels of TNFα mRNA compared with WT cells. This was accompanied by higher levels of p-p65, a down-stream target of TNFα, suggesting that BMSCs from Col6α2-KO mice are highly sensitive to TNFα signaling. Taken together, our data imply that Col6a2 deficiency causes trabecular bone loss by enhancing osteoclast differentiation through enhanced TNFα signaling.
Collapse
Affiliation(s)
- Hai T Pham
- Molecular Biology of Bones and Teeth Section, Department of Health and Human Services (DHHS), National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Building 30 Room 5A509, Bethesda, MD, 20892, USA
| | - Vardit Kram
- Molecular Biology of Bones and Teeth Section, Department of Health and Human Services (DHHS), National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Building 30 Room 5A509, Bethesda, MD, 20892, USA
| | - Qurratul-Ain Dar
- Molecular Biology of Bones and Teeth Section, Department of Health and Human Services (DHHS), National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Building 30 Room 5A509, Bethesda, MD, 20892, USA
| | - Taishi Komori
- Molecular Biology of Bones and Teeth Section, Department of Health and Human Services (DHHS), National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Building 30 Room 5A509, Bethesda, MD, 20892, USA
| | - Youngmi Ji
- Molecular Biology of Bones and Teeth Section, Department of Health and Human Services (DHHS), National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Building 30 Room 5A509, Bethesda, MD, 20892, USA
| | - Payam Mohassel
- Neuromuscular and Neurogenic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stoke, Department of Health and Human Services, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jachinta Rooney
- Neuromuscular and Neurogenic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stoke, Department of Health and Human Services, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Li Li
- Molecular Biology of Bones and Teeth Section, Department of Health and Human Services (DHHS), National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Building 30 Room 5A509, Bethesda, MD, 20892, USA
| | - Tina M Kilts
- Molecular Biology of Bones and Teeth Section, Department of Health and Human Services (DHHS), National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Building 30 Room 5A509, Bethesda, MD, 20892, USA
| | - Carsten Bonnemann
- Neuromuscular and Neurogenic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stoke, Department of Health and Human Services, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Shireen Lamande
- Department of Pediatrics, University of Melbourne, Parkville, Australia
| | - Marian F Young
- Molecular Biology of Bones and Teeth Section, Department of Health and Human Services (DHHS), National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Building 30 Room 5A509, Bethesda, MD, 20892, USA.
| |
Collapse
|
45
|
Rezende TMB, Ribeiro Sobrinho AP, Vieira LQ, Sousa MGDC, Kawai T. Mineral trioxide aggregate (MTA) inhibits osteoclastogenesis and osteoclast activation through calcium and aluminum activities. Clin Oral Investig 2020; 25:1805-1814. [PMID: 32789653 DOI: 10.1007/s00784-020-03483-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/29/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To evaluate the effect(s) of mineral trioxide aggregate (MTA) on in vitro RANKL-mediated osteoclast-dependent bone resorption events and the influence of Ca2+ and Al3+ on the osteoclastogenesis inhibition by MTA. MATERIALS AND METHODS Two types of osteoclast precursors, RAW 264.7 (RAW) cell line or bone marrow cells (obtained from BALB/c mice and stimulated with recombinant (r) macrophage colony stimulation factor (M-CSF), were stimulated with or without recombinant (r) activator of nuclear kappa B ligand (RANKL), in the presence or absence of MTA for 6 to 8 days. White Angelus MTA and Bios MTA (Angelus, Londrina, Paraná, Brazil) were prepared and inserted into capillary tubes (direct contact surface = 0.50 mm2 and 0.01 mm2). Influence of MTA on these types of osteoclast precursors was measured by the number of differentiated tartrate-resistant acid phosphatase (TRAP)-positive multinuclear cells (RAW and bone marrow cells), TRAP enzyme activity (RAW cells), cathepsin K gene expression (RAW cells), and resorptive pit formation (RAW cells) by mature osteoclasts. Besides, RAW cells were also stimulated with Ca2+ and Al3+ to evaluate the influence of these ions on MTA anti-osteoclastogenic potential. RESULTS In bone marrow and RAW cells, the number of TRAP-positive mature osteoclast cells induced by rRANKL was significantly inhibited by the presence of MTA compared with control rRANKL stimulation without MTA (p < 0.05), along with the reduction of TRAP enzyme activity (p < 0.05) and the low expression of cathepsin K gene (p < 0.05). In contrast, to control mature osteoclasts, the resorption area on dentin was significantly decreased for mature osteoclasts incubated with MTA (p < 0.05). rRANKL-stimulated RAW cells treated with Ca2+ and Al3+ decreased the number of osteoclasts cells. Besides, the aluminum oxide was the dominant suppressor of the osteoclastogenesis process. CONCLUSIONS MTA significantly suppressed RANKL-mediated osteoclastogenesis and osteoclast activity and, therefore, appears able to suppress bone resorption events in periapical lesions. This process might be related to Ca2+ and Al3+ activities. CLINICAL RELEVANCE MTA is an important worldwidely acknowleged biomaterial. The knowledge about its molecular activities on osteoclasts might contribute to improving the understanding of its clinical efficacy.
Collapse
Affiliation(s)
- Taia Maria Berto Rezende
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil. .,Curso de Odontologia, Universidade Católica de Brasília, Brasília, DF, Brazil. .,Programa de Pós-graduação em Ciências da Saúde, Universidade de Brasília, Brasília, DF, Brazil.
| | - Antônio Paulino Ribeiro Sobrinho
- Departamento de Odontologia Restauradora, Faculdade de Odontologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Leda Quercia Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Toshihisa Kawai
- Department of Oral Science and Translational Research, College of Dental Medicine, NOVA Southeastern University, Fort Lauderdale, FL, USA.,Cell Therapy Institute, Center for Collaborative Research, NOVA Southeastern University, Fort Lauderdale, FL, USA
| |
Collapse
|
46
|
Wang M, Xia F, Wei Y, Wei X. Molecular mechanisms and clinical management of cancer bone metastasis. Bone Res 2020; 8:30. [PMID: 32793401 PMCID: PMC7391760 DOI: 10.1038/s41413-020-00105-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/03/2019] [Accepted: 10/23/2019] [Indexed: 02/05/2023] Open
Abstract
As one of the most common metastatic sites of malignancies, bone has a unique microenvironment that allows metastatic tumor cells to grow and flourish. The fenestrated capillaries in the bone, bone matrix, and bone cells, including osteoblasts and osteoclasts, together maintain the homeostasis of the bone microenvironment. In contrast, tumor-derived factors act on bone components, leading to subsequent bone resorption or excessive bone formation. The various pathways involved also provide multiple targets for therapeutic strategies against bone metastases. In this review, we summarize the current understanding of the mechanism of bone metastases. Based on the general process of bone metastases, we specifically highlight the complex crosstalk between tumor cells and the bone microenvironment and the current management of cancer bone metastases.
Collapse
Affiliation(s)
- Manni Wang
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan P.R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| |
Collapse
|
47
|
Eraković M, Duka M, Bekić M, Tomić S, Ismaili B, Vučević D, Čolić M. Anti‐inflammatory and immunomodulatory effects of Biodentine on human periapical lesion cells in culture. Int Endod J 2020; 53:1398-1412. [DOI: 10.1111/iej.13351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/19/2022]
Affiliation(s)
- M. Eraković
- Clinic for Stomatology Military Medical Academy BelgradeSerbia
| | - M. Duka
- Clinic for Stomatology Military Medical Academy BelgradeSerbia
| | - M. Bekić
- Institute for the Application of Nuclear Energy Zemun Serbia
| | - S. Tomić
- Institute for the Application of Nuclear Energy Zemun Serbia
| | - B. Ismaili
- Polyclinic, Ismaili Gostivar North Macedonia
| | - D. Vučević
- Medical Faculty of the Military Medical Academy University of Defense Belgrade Serbia
| | - M. Čolić
- Institute for the Application of Nuclear Energy Zemun Serbia
- Medical Faculty of the Military Medical Academy University of Defense Belgrade Serbia
- Medical Faculty Foča University of East Sarajevo R.Srpska Bosnia and Herzegovina
| |
Collapse
|
48
|
Dimethyl fumarate prevents osteoclastogenesis by decreasing NFATc1 expression, inhibiting of erk and p38 MAPK phosphorylation, and suppressing of HMGB1 release. Biochem Biophys Res Commun 2020; 530:455-461. [PMID: 32553625 DOI: 10.1016/j.bbrc.2020.05.088] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022]
Abstract
Osteoclasts are multinucleated bone-resorbing cells derived from monocyte/macrophage progenitor cells. Excessive formation and resorbing activities of osteoclasts are involved in the bone-destructive pathologies of rheumatoid arthritis and osteoporosis. Recently, it has been found that nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor for anti-oxidative stress genes, functions in osteoclastogenesis. Dimethyl fumarate (DMF) is a potent activator of Nrf2 and has been shown to inhibit osteoclastogenesis. Here, we investigated the mechanisms of this inhibition by examining the activation of several signalling pathways during the differentiation of bone marrow-derived macrophages into osteoclasts. DMF inhibited the differentiation of osteoclasts in a dose-dependent manner and suppressed the bone-resorbing activity of osteoclasts. DMF treatment decreased the expression of nuclear factor of activated T-cells cytoplasmic-1, and significantly decreased phosphorylation of extracellular signal-regulated kinase and p38 mitogen-activated protein kinase in osteoclasts. We also found that DMF inhibited the extracellular release of high mobility group box 1, associated with an up-regulation of heme oxygenase-1, likely mediated through Nrf2 activation. Our results indicate that DMF inhibits osteoclast differentiation through multiple pathways.
Collapse
|
49
|
Strisciuglio C, Scarpato E, Cenni S, Serra MR, Giugliano FP, Mainolfi CG, Dolce P, Martinelli M, Staiano A, Miele E. Improvement of body composition and bone mineral density after enteral nutrition in pediatric Crohn disease. Dig Liver Dis 2020; 52:630-636. [PMID: 32273172 DOI: 10.1016/j.dld.2020.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/26/2020] [Accepted: 03/04/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The relationship between exclusive enteral nutrition (EEN) and bone status is poorly defined in pediatric Crohn disease (CD). AIMS The aim of this study was to investigate the impact of EEN on body composition, nutritional status, and bone mineral density (BMD) in an incident CD cohort. METHODS 18 newly diagnosed CD children starting EEN for 8 weeks were prospectively enrolled and evaluated at baseline and after 8 (T8), 26 (T26) and 52 weeks (T52) from diagnosis. The Fat Free Mass (FFM) and the Resting Energy Expenditure (REE) were measured through Bioelectrical Impedance (BIA) and the BMD was assessed by dual-energy X-ray (DXA). We compared DXA data of IBD patients to the data obtained in 15 healthy controls. RESULTS CD children had a significant lower BMD compared to healthy control both at baseline (p<0.0001), and after EEN therapy at T52 (p=0.0004); although at this latest time point CD children had a significant increase of BMD compared to baseline (p=0.0015). The BIA analysis showed a significant increase at T26 and T52 of FFM and REE. T52. FFM measured by BIA and BMD measured by DXA were significantly correlated. CONCLUSION EEN improves nutritional status and bone mineral composition.
Collapse
Affiliation(s)
- C Strisciuglio
- Department of Woman, Child and General and Specialized Surgery, University of the Campania "Luigi Vanvitelli", Naples, Italy
| | - E Scarpato
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - S Cenni
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - M R Serra
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - F P Giugliano
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - C G Mainolfi
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - P Dolce
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - M Martinelli
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - A Staiano
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples, Italy.
| | - E Miele
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
50
|
Duan L, Zuo J, Zhang F, Li B, Xu Z, Zhang H, Yang B, Song W, Jiang J. Magnetic Targeting of HU-MSCs in the Treatment of Glucocorticoid-Associated Osteonecrosis of the Femoral Head Through Akt/Bcl2/Bad/Caspase-3 Pathway. Int J Nanomedicine 2020; 15:3605-3620. [PMID: 32547017 PMCID: PMC7247730 DOI: 10.2147/ijn.s244453] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 04/30/2020] [Indexed: 01/26/2023] Open
Abstract
Purpose Osteonecrosis of the femoral head (ONFH) is a chronic and irreversible disease that eventually develops into a joint collapse and results in joint dysfunction. Early intervention and treatment are essential for preserving the joints and avoiding hip replacement. In this study, a system of human umbilical mesenchymal stem cells-supermagnetic iron oxide nanoparticles (NPs) @polydopamine (SCIOPs) was constructed. The magnetic targeting system gathers in the lesion area, inhibits the apoptosis of bone cells, enhances osteogenic effect, and effectively treats ONFH under external magnetic field. Materials and Methods The supermagnetic iron oxide NPs @polydopamine (SPION@PDA NPs) were characterized by transmission electron microscopy and zeta potential, respectively. The effects of SPION@PDA NPs on the viability, proliferation, and differentiation of stem cells were detected by the CCK8 method, flow cytometry, and staining, respectively. The serum inflammatory indicators were detected by Luminex method. The bone mass of the femoral head was analyzed by micro computed tomography. The expression of apoptosis and osteoblast-related cytokines was detected by Western blotting. The osteogenesis of the femoral head was detected by histological and immunohistochemical sections. Results The SCIOPs decreased the pro-inflammatory factors, and the micro CT showed that the bone repair of the femoral head was enhanced after treatment. The hematoxylin and eosin sections also showed an increase in the osteogenesis in the femoral head. Western blotting results showed and increased expression of anti-apoptotic proteins Akt and Bcl-2, decreased expression of apoptotic proteins caspase-3 and Bad, and increased expression of osteogenic proteins Runx-2 and Osterix in the femoral head. Conclusion Under the effect of magnetic field and homing ability of stem cells, SCIOPs inhibited the apoptosis of osteoblasts, improved the proliferation ability of osteoblasts, and promoted bone repair in the femoral head through the Akt/Bcl-2/Bad/caspase-3 signaling pathway, thereby optimizing the tissue repair ability.
Collapse
Affiliation(s)
- Lian Duan
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Jianlin Zuo
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Fuqiang Zhang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Binxi Li
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, Jilin, People's Republic of China
| | - Zhonghang Xu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, Jilin, People's Republic of China
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, Jilin, People's Republic of China
| | - Wenzhi Song
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|