1
|
Matsuki Y, Nakamura T. Spondyloarthritis and Tietze's syndrome: A re-evaluation. Mod Rheumatol 2024; 35:1-6. [PMID: 39271145 DOI: 10.1093/mr/roae086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/16/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024]
Abstract
Spondyloarthritis (SpA) comprises a number of related but different disorders with distinct phenotypes: psoriatic arthritis, reactive arthritis, arthritis related to inflammatory bowel disease, undifferentiated arthritis, and ankylosing spondylitis (the well-known prototypic subtype). Differentiating rheumatic diseases, such as rheumatoid arthritis, synovitis-acne-pustulosis-hyperostosis-osteitis syndrome, pustulotic arthro-osteitis, gout, and SpA, is difficult because they all may manifest swelling at the upper anterior chest wall, often involve the sternocostal and/or sternoclavicular joints, and clearly show cutaneous nodular symptoms, so that they may mimic Tietze's syndrome in the presentation. Tietze's syndrome is a benign, self-limiting entity with tender, non-suppurative swelling in the upper anterior chest wall, especially at the second and third costosternal junctions and the sternoclavicular joint. Therefore, distinguishing SpA from Tietze's syndrome during an entire disease course is important, even after an initial tentative diagnosis. This article aims to re-evaluate the importance of Tietze's syndrome in the differential diagnosis of SpA, while keeping in mind information about rheumatic diseases affecting the upper anterior chest wall.
Collapse
Affiliation(s)
- Yasunori Matsuki
- Section of Rheumatology, Internal Medicine, Sakurajyuji Yatsushiro Rehabilitation Hospital, Yatsushiro, Kumamoto, Japan
| | - Tadashi Nakamura
- Division of Clinical Rheumatology, Sakurajyuji Hospital, Kumamoto, Japan
| |
Collapse
|
2
|
Dong Q, Zhou J, Feng M, Kong L, Fang B, Zhang Z. A review of bacterial and osteoclast differentiation in bone infection. Microb Pathog 2024; 197:107102. [PMID: 39505086 DOI: 10.1016/j.micpath.2024.107102] [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: 03/29/2024] [Revised: 10/18/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
Bone infections are characterized by bacterial invasion of the bone microenvironment and subsequent bone structure deterioration. This holds significance because osteoclasts, which are the only cells responsible for bone resorption, are abnormally stimulated during bone infections. Multiple communication factors secreted by bone stromal cells regulate the membrane of osteoclast progenitor cells, thereby maintaining bone homeostasis through the expression of many types of receptors. During infection, the immunoinflammatory response triggered by bacterial invasion and multiple virulence factors of bacterial origin can disrupt osteoclast homeostasis. Therefore, clarifying the pathways through which bacteria affect osteoclasts can offer a theoretical basis for preventing and treating bone infections. This review summarizes studies investigating bone destruction caused by different bacterial infections. In conclusion, bacteria can affect osteoclast metabolic activity through multiple pathways, including direct contact, release of virulence factors, induction of immunoinflammatory responses, influence on bone stromal cell metabolism, and intracellular infections.
Collapse
Affiliation(s)
- Qi Dong
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Jiuqin Zhou
- Department of Infectious Disease of Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Mingzhe Feng
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Lingqiang Kong
- Department of Orthopedics, the Central Hospital Affiliated to Shaoxing University, Shaoxing, 312030, China.
| | - Bin Fang
- Department of Orthopedics, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310000, China.
| | - Zhen Zhang
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China.
| |
Collapse
|
3
|
Ren J, Kitaura H, Noguchi T, Ohori F, Marahleh A, Ma J, Kanou K, Fan Z, Mizoguchi I. Exogenous Angiotensin-(1-7) Provides Protection Against Inflammatory Bone Resorption and Osteoclastogenesis by Inhibition of TNF-α Expression in Macrophages. Calcif Tissue Int 2024; 115:432-444. [PMID: 39030433 PMCID: PMC11405502 DOI: 10.1007/s00223-024-01257-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 07/11/2024] [Indexed: 07/21/2024]
Abstract
Renin-angiotensin-aldosterone system plays a crucial role in the regulation of blood pressure and fluid homeostasis. It is reported to be involved in mediating osteoclastogenesis and bone loss in diseases of inflammatory bone resorption such as osteoporosis. Angiotensin-(1-7), a product of Angiotensin I and II (Ang I, II), is cleaved by Angiotensin-converting enzyme 2 and then binds to Mas receptor to counteract inflammatory effects produced by Ang II. However, the mechanism by which Ang-(1-7) reduces bone resorption remains unclear. Therefore, we aim to elucidate the effects of Ang-(1-7) on lipopolysaccharide (LPS)-induced osteoclastogenesis. In vivo, mice were supracalvarial injected with Ang-(1-7) or LPS ± Ang-(1-7) subcutaneously. Bone resorption and osteoclast formation were compared using micro-computed tomography, tartrate-resistant acid phosphatase (TRAP) stain, and real-time PCR. We found that Ang-(1-7) attenuated tumor necrosis factor (TNF)-α, TRAP, and Cathepsin K expression from calvaria and decreased osteoclast number along with bone resorption at the suture mesenchyme. In vitro, RANKL/TNF-α ± Ang-(1-7) was added to cultures of bone marrow-derived macrophages (BMMs) and osteoclast formation was measured via TRAP staining. The effect of Ang-(1-7) on LPS-induced osteoblasts RANKL expression and peritoneal macrophages TNF-α expression was also investigated. The effect of Ang-(1-7) on the MAPK and NF-κB pathway was studied by Western blotting. As a result, Ang-(1-7) reduced LPS-stimulated macrophages TNF-α expression and inhibited the MAPK and NF-κB pathway activation. However, Ang-(1-7) did not affect osteoclastogenesis induced by RANKL/TNF-α nor reduce osteoblasts RANKL expression in vitro. In conclusion, Ang-(1-7) alleviated LPS-induced osteoclastogenesis and bone resorption in vivo via inhibiting TNF-α expression in macrophages.
Collapse
Affiliation(s)
- Jiayi Ren
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Hideki Kitaura
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan.
| | - Takahiro Noguchi
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Fumitoshi Ohori
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Aseel Marahleh
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | - Jinghan Ma
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Kayoko Kanou
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Ziqiu Fan
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Itaru Mizoguchi
- Department of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
4
|
Riyaz S, Sun Y, Helmholz H, Medina TP, Medina OP, Wiese B, Will O, Albaraghtheh T, Mohamad FH, Hövener JB, Glüer CC, Römer RW. Inflammatory response toward a Mg-based metallic biomaterial implanted in a rat femur fracture model. Acta Biomater 2024; 185:41-54. [PMID: 38969080 DOI: 10.1016/j.actbio.2024.06.040] [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: 03/12/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024]
Abstract
The immune system plays an important role in fracture healing, by modulating the pro-inflammatory and anti-inflammatory responses occurring instantly upon injury. An imbalance in these responses can lead to adverse outcomes, such as non-union of fractures. Implants are used to support and stabilize complex fractures. Biodegradable metallic implants offer the potential to avoid a second surgery for implant removal, unlike non-degradable implants. However, considering our dynamic immune system it is important to conduct in-depth studies on the immune response to these implants in living systems. In this study, we investigated the immune response to Mg and Mg-10Gd in vivo in a rat femur fracture model with external fixation. In vivo imaging using liposomal formulations was used to monitor the fluorescence-related inflammation over time. We combine ex vivo methods with our in vivo study to evaluate and understand the systemic and local effects of the implants on the immune response. We observed no significant local or systemic effects in the Mg-10Gd implanted group compared to the SHAM and Mg implanted groups over time. Our findings suggest that Mg-10Gd is a more compatible implant material than Mg, with no adverse effects observed in the early phase of fracture healing during our 4-week study. STATEMENT OF SIGNIFICANCE: Degradable metallic implants in form of Mg and Mg-10Gd intramedullary pins were assessed in a rat femur fracture model, alongside a non-implanted SHAM group with special respect to the potential to induce an inflammatory response. This pre-clinical study combines innovative non-invasive in vivo imaging techniques associated with multimodal, ex vivo cellular and molecular analytics. The study contributes to the development and evaluation of degradable biometals and their clinical application potential. The study results indicate that Mg-10Gd did not exhibit any significant harmful effects compared to the SHAM and Mg groups.
Collapse
Affiliation(s)
- Sana Riyaz
- Helmholtz-Zentrum hereon GmbH, Institute of Metallic Biomaterials, Max-Planck-Straße 1, Geesthacht 21502, Germany.
| | - Yu Sun
- Helmholtz-Zentrum hereon GmbH, Institute of Metallic Biomaterials, Max-Planck-Straße 1, Geesthacht 21502, Germany
| | - Heike Helmholz
- Helmholtz-Zentrum hereon GmbH, Institute of Metallic Biomaterials, Max-Planck-Straße 1, Geesthacht 21502, Germany.
| | - Tuula Penate Medina
- Section Biomedical Imaging, Department of Radiology and Neuroradiology University Hospital Schleswig-Holstein Campus Kiel, Kiel University, Am Botanischen Garten 14, 24118 Kiel, Germany; Institute for Experimental Cancer Research, Kiel University, 24105 Kiel, Germany
| | - Oula Penate Medina
- Section Biomedical Imaging, Department of Radiology and Neuroradiology University Hospital Schleswig-Holstein Campus Kiel, Kiel University, Am Botanischen Garten 14, 24118 Kiel, Germany; Institute for Experimental Cancer Research, Kiel University, 24105 Kiel, Germany; Lonza Netherlands B.V., 6167 RB Geleen, the Netherlands
| | - Björn Wiese
- Helmholtz-Zentrum hereon GmbH, Institute of Metallic Biomaterials, Max-Planck-Straße 1, Geesthacht 21502, Germany
| | - Olga Will
- Section Biomedical Imaging, Department of Radiology and Neuroradiology University Hospital Schleswig-Holstein Campus Kiel, Kiel University, Am Botanischen Garten 14, 24118 Kiel, Germany
| | - Tamadur Albaraghtheh
- Helmholtz-Zentrum hereon GmbH, Institute of Metallic Biomaterials, Max-Planck-Straße 1, Geesthacht 21502, Germany; Helmholtz-Zentrum hereon GmbH, Institute of Surface Science, Max-Planck-Straße 1, Geesthacht 21502, Germany
| | - Farhad Haj Mohamad
- Section Biomedical Imaging, Department of Radiology and Neuroradiology University Hospital Schleswig-Holstein Campus Kiel, Kiel University, Am Botanischen Garten 14, 24118 Kiel, Germany
| | - Jan-Bernd Hövener
- Section Biomedical Imaging, Department of Radiology and Neuroradiology University Hospital Schleswig-Holstein Campus Kiel, Kiel University, Am Botanischen Garten 14, 24118 Kiel, Germany
| | - Claus Christian Glüer
- Section Biomedical Imaging, Department of Radiology and Neuroradiology University Hospital Schleswig-Holstein Campus Kiel, Kiel University, Am Botanischen Garten 14, 24118 Kiel, Germany
| | - Regine Willumeit Römer
- Helmholtz-Zentrum hereon GmbH, Institute of Metallic Biomaterials, Max-Planck-Straße 1, Geesthacht 21502, Germany
| |
Collapse
|
5
|
Zhang Z, Wu W, Li M, Du L, Li J, Yin X, Zhang W. Mesenchymal stem cell–derived extracellular vesicles: A novel nanoimmunoregulatory tool in musculoskeletal diseases. NANO TODAY 2024; 57:102343. [DOI: 10.1016/j.nantod.2024.102343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Yue Y, Dai W, Wei Y, Cao S, Liao S, Li A, Liu P, Lin J, Zeng H. Unlocking the potential of exosomes: a breakthrough in the theranosis of degenerative orthopaedic diseases. Front Bioeng Biotechnol 2024; 12:1377142. [PMID: 38699435 PMCID: PMC11064847 DOI: 10.3389/fbioe.2024.1377142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/01/2024] [Indexed: 05/05/2024] Open
Abstract
Degenerative orthopaedic diseases pose a notable worldwide public health issue attributable to the global aging population. Conventional medical approaches, encompassing physical therapy, pharmaceutical interventions, and surgical methods, face obstacles in halting or reversing the degenerative process. In recent times, exosome-based therapy has gained widespread acceptance and popularity as an effective treatment for degenerative orthopaedic diseases. This therapeutic approach holds the potential for "cell-free" tissue regeneration. Exosomes, membranous vesicles resulting from the fusion of intracellular multivesicles with the cell membrane, are released into the extracellular matrix. Addressing challenges such as the rapid elimination of natural exosomes in vivo and the limitation of drug concentration can be effectively achieved through various strategies, including engineering modification, gene overexpression modification, and biomaterial binding. This review provides a concise overview of the source, classification, and preparation methods of exosomes, followed by an in-depth analysis of their functions and potential applications. Furthermore, the review explores various strategies for utilizing exosomes in the treatment of degenerative orthopaedic diseases, encompassing engineering modification, gene overexpression, and biomaterial binding. The primary objective is to provide a fresh viewpoint on the utilization of exosomes in addressing bone degenerative conditions and to support the practical application of exosomes in the theranosis of degenerative orthopaedic diseases.
Collapse
Affiliation(s)
- Yaohang Yue
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wei Dai
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yihao Wei
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Siyang Cao
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Shuai Liao
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Aikang Li
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Peng Liu
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jianjing Lin
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Hui Zeng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Du J, Wang Y, Wu C, Zhang X, Zhang X, Xu X. Targeting bone homeostasis regulation: potential of traditional Chinese medicine flavonoids in the treatment of osteoporosis. Front Pharmacol 2024; 15:1361864. [PMID: 38628649 PMCID: PMC11018902 DOI: 10.3389/fphar.2024.1361864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024] Open
Abstract
Osteoporosis is a systemic metabolic disease characterized by disrupted bone formation/resorption and homeostasis. Flavonoids extracted from traditional Chinese medicinal plants regulate bone homeostasis by intervening in differentiating bone marrow mesenchymal stem cells, balancing the bone immune system, inhibiting oxidative stress response, and reversing iron overload. The target molecules and signaling pathways, such as Wnt/β-catenin and OPG/RANKL/RANK, directly affect osteoblast/osteoclast activity, exhibiting significant potential in the treatment of OP. Therefore, this study presents a systematic review of the recent literature to provide comprehensive information on the traditional Chinese medicine flavonoids involved in the regulation of bone homeostasis. Also, the molecular mechanisms and pharmacological uses of these metabolites are summarized, and their clinical translation and development potential are discussed.
Collapse
Affiliation(s)
- Jiazhe Du
- Graduate School, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yincang Wang
- Graduate School, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Chengliang Wu
- Institute of Orthopedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyu Zhang
- Graduate School, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xiaofeng Zhang
- Teaching and Research Section of Orthopedics and Traumatology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xilin Xu
- Department of Orthopedics, The Third Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
8
|
Zhang B, He Z, Guo J, Li F, Huang Z, Zheng W, Xing W, Li M, Zhu Y, Yang X. Sesamin-mediated high expression of BECN2 ameliorates cartilage endplate degeneration by reducing autophagy and inflammation. Aging (Albany NY) 2024; 16:1145-1160. [PMID: 38284902 PMCID: PMC10866406 DOI: 10.18632/aging.205386] [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: 07/29/2023] [Accepted: 11/13/2023] [Indexed: 01/30/2024]
Abstract
Lumbar disc degeneration (LDD) is a prevalent clinical spinal disease characterized by the calcification and degeneration of the cartilage endplate (CEP), which significantly reduces nutrient supply to the intervertebral disc. Traditional Chinese medicine offers a conservative and effective approach for treating LDD. We aimed to investigate the molecular mechanisms underlying the therapeutic effects of Sesamin in LDD treatment. Transcriptome sequencing was used to analyze the effect of Sesamin on LPS-induced ATDC5. We explored the role of BECN2, a target gene of Sesamin, in attenuating LPS-induced degeneration of ATDC5 cells. Our results revealed the identification of 117 differentially expressed genes (DEGs), with 54 up-regulated and 63 down-regulated genes. Notably, Sesamin significantly increased the expression of BECN2 in LPS-induced ATDC5 cell degeneration. Overexpressed BECN2 enhanced cell viability and inhibited cell apoptosis in LPS-induced ATDC5 cells, while BECN2 knockdown reduced cell viability and increased apoptosis. Furthermore, BECN2 played a crucial role in attenuating chondrocyte degeneration by modulating autophagy and inflammation. Specifically, BECN2 suppressed autophagy by reducing the expression of ATG14, VPS34, and GASP1, and alleviated the inflammatory response by decreasing the expression of inflammasome proteins NLRP3, NLRC4, NLRP1, and AIM2. In vivo experiments further supported the beneficial effects of Sesamin in mitigating LDD. This study provides novel insights into the potential molecular mechanism of Sesamin in treating LDD, highlighting its ability to mediate autophagy and inflammation inhibition via targeting the BECN2. This study provides a new therapeutic strategy for the treatment of LDD, as well as a potential molecular target for LDD.
Collapse
Affiliation(s)
- Baining Zhang
- Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Zhiwei He
- Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Jialin Guo
- Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Feng Li
- Department of Spine Surgery, Area A, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Zhi Huang
- Department of Spine Surgery, Area A, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Wenkai Zheng
- Department of Spine Surgery, Area A, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Wenhua Xing
- Department of Spine Surgery, Area A, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Manglai Li
- Department of Bone and Soft Tissue Oncology, The Affiliated People’s Hospital of inner Mongolia Medical University, Peking University Cancer Hospital, Hohhot, Inner Mongolia, China
| | - Yong Zhu
- Department of Bone and Soft Tissue Oncology, The Affiliated People’s Hospital of inner Mongolia Medical University, Peking University Cancer Hospital, Hohhot, Inner Mongolia, China
| | - Xuejun Yang
- Department of Spine Surgery, Area A, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
9
|
Panez-Toro I, Heymann D, Gouin F, Amiaud J, Heymann MF, Córdova LA. Roles of inflammatory cell infiltrate in periprosthetic osteolysis. Front Immunol 2023; 14:1310262. [PMID: 38106424 PMCID: PMC10722268 DOI: 10.3389/fimmu.2023.1310262] [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: 10/09/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
Classically, particle-induced periprosthetic osteolysis at the implant-bone interface has explained the aseptic loosening of joint replacement. This response is preceded by triggering both the innate and acquired immune response with subsequent activation of osteoclasts, the bone-resorbing cells. Although particle-induced periprosthetic osteolysis has been considered a foreign body chronic inflammation mediated by myelomonocytic-derived cells, current reports describe wide heterogeneous inflammatory cells infiltrating the periprosthetic tissues. This review aims to discuss the role of those non-myelomonocytic cells in periprosthetic tissues exposed to wear particles by showing original data. Specifically, we discuss the role of T cells (CD3+, CD4+, and CD8+) and B cells (CD20+) coexisting with CD68+/TRAP- multinucleated giant cells associated with both polyethylene and metallic particles infiltrating retrieved periprosthetic membranes. This review contributes valuable insight to support the complex cell and molecular mechanisms behind the aseptic loosening theories of orthopedic implants.
Collapse
Affiliation(s)
- Isidora Panez-Toro
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Chile, Independencia, Santiago, Chile
- Nantes Université, Centre National de Recherche Scientifique (CNRS), UMR6286, US2B, Nantes, France
- Institut de Cancérologie de l’Ouest, Tumor Heterogeneity and Precision Medicine Laboratory, Saint-Herblain, France
| | - Dominique Heymann
- Nantes Université, Centre National de Recherche Scientifique (CNRS), UMR6286, US2B, Nantes, France
- Institut de Cancérologie de l’Ouest, Tumor Heterogeneity and Precision Medicine Laboratory, Saint-Herblain, France
- Nantes Université, Laboratory of Histology and Embryology, Medical School, Nantes, France
- The University of Sheffield, Dept of Oncology and Metabolism, Sheffield, United Kingdom
| | - François Gouin
- Department of Surgical Oncology, Centre Léon Bérard, Lyon, France
| | - Jérôme Amiaud
- Nantes Université, Laboratory of Histology and Embryology, Medical School, Nantes, France
| | - Marie-Françoise Heymann
- Nantes Université, Centre National de Recherche Scientifique (CNRS), UMR6286, US2B, Nantes, France
- Institut de Cancérologie de l’Ouest, Tumor Heterogeneity and Precision Medicine Laboratory, Saint-Herblain, France
| | - Luis A. Córdova
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, University of Chile, Independencia, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Oral and Maxillofacial Surgery, Clínica MEDS, Santiago, Chile
| |
Collapse
|
10
|
Zhang Z, Ji C, Wang D, Wang M, She X, Song D, Xu X, Zhang D. Maresin1: A multifunctional regulator in inflammatory bone diseases. Int Immunopharmacol 2023; 120:110308. [PMID: 37192551 DOI: 10.1016/j.intimp.2023.110308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/18/2023]
Abstract
Inflammation plays a crucial role in the physical response to danger signals, the elimination of toxic stimuli, and the restoration of homeostasis. However, dysregulated inflammatory responses lead to tissue damage, and chronic inflammation can disrupt osteogenic-osteoclastic homeostasis, ultimately leading to bone loss. Maresin1 (MaR1), a member of the specialized pro-resolving mediators (SPMs) family, has been found to possess significant anti-inflammatory, anti-allergic, pro-hemolytic, pro-healing, and pain-relieving properties. MaR1 is synthesized by macrophages (Mφs) and omega-3 fatty acids, and it may have the potential to promote bone homeostasis and treat inflammatory bone diseases. MaR1 has been found to stimulate osteoblast proliferation through leucine-rich repeat G protein-coupled receptor 6 (LGR6). It also activates Mφ phagocytosis and M2-type polarization, which helps to control the immune system. MaR1 can regulate T cells to exert anti-inflammatory effects and inhibit neutrophil infiltration and recruitment. In addition, MaR1 is involved in antioxidant signaling, including nuclear factor erythroid 2-related factor 2 (NRF2). It has also been found to promote the autophagic behavior of periodontal ligament stem cells, stimulate Mφs against pathogenic bacteria, and regulate tissue regeneration and repair. In summary, this review provides new information and a comprehensive overview of the critical roles of MaR1 in inflammatory bone diseases, indicating its potential as a therapeutic approach for managing skeletal metabolism and inflammatory bone diseases.
Collapse
Affiliation(s)
- Zhanwei Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Chonghao Ji
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | | | - Maoshan Wang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Xiao She
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Dawei Song
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xin Xu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| | - Dongjiao Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| |
Collapse
|
11
|
Ke Q, Greenawalt AN, Manukonda V, Ji X, Tisch RM. The regulation of self-tolerance and the role of inflammasome molecules. Front Immunol 2023; 14:1154552. [PMID: 37081890 PMCID: PMC10110889 DOI: 10.3389/fimmu.2023.1154552] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/17/2023] [Indexed: 04/07/2023] Open
Abstract
Inflammasome molecules make up a family of receptors that typically function to initiate a proinflammatory response upon infection by microbial pathogens. Dysregulation of inflammasome activity has been linked to unwanted chronic inflammation, which has also been implicated in certain autoimmune diseases such as multiple sclerosis, rheumatoid arthritis, type 1 diabetes, systemic lupus erythematosus, and related animal models. Classical inflammasome activation-dependent events have intrinsic and extrinsic effects on both innate and adaptive immune effectors, as well as resident cells in the target tissue, which all can contribute to an autoimmune response. Recently, inflammasome molecules have also been found to regulate the differentiation and function of immune effector cells independent of classical inflammasome-activated inflammation. These alternative functions for inflammasome molecules shape the nature of the adaptive immune response, that in turn can either promote or suppress the progression of autoimmunity. In this review we will summarize the roles of inflammasome molecules in regulating self-tolerance and the development of autoimmunity.
Collapse
Affiliation(s)
- Qi Ke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ashley Nicole Greenawalt
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Veera Manukonda
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Xingqi Ji
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Roland Michael Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- *Correspondence: Roland Michael Tisch,
| |
Collapse
|
12
|
Kim CH. Complex regulatory effects of gut microbial short-chain fatty acids on immune tolerance and autoimmunity. Cell Mol Immunol 2023; 20:341-350. [PMID: 36854801 PMCID: PMC10066346 DOI: 10.1038/s41423-023-00987-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/10/2023] [Indexed: 03/02/2023] Open
Abstract
Immune tolerance deletes or suppresses autoreactive lymphocytes and is established at multiple levels during the development, activation and effector phases of T and B cells. These mechanisms are cell-intrinsically programmed and critical in preventing autoimmune diseases. We have witnessed the existence of another type of immune tolerance mechanism that is shaped by lifestyle choices, such as diet, microbiome and microbial metabolites. Short-chain fatty acids (SCFAs) are the most abundant microbial metabolites in the colonic lumen and are mainly produced by the microbial fermentation of prebiotics, such as dietary fiber. This review focuses on the preventive and immunomodulatory effects of SCFAs on autoimmunity. The tissue- and disease-specific effects of dietary fiber, SCFAs and SCFA-producing microbes on major types of autoimmune diseases, including type I diabetes, multiple sclerosis, rheumatoid arthritis and lupus, are discussed. Additionally, their key regulatory mechanisms for lymphocyte development, tissue barrier function, host metabolism, immunity, autoantibody production, and inflammatory effector and regulatory lymphocytes are discussed. The shared and differential effects of SCFAs on different types and stages of autoimmune diseases are discussed.
Collapse
Affiliation(s)
- Chang H Kim
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA.
- Mary H. Weiser Food Allergy Center, Center for Gastrointestinal Research, and Rogel Center for Cancer Research, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
13
|
Han Y, Yang H, Hua Z, Nie S, Xu S, Zhou C, Chen F, Li M, Yu Q, Sun Y, Wei Y, Wang X. Rotating Magnetic Field Mitigates Ankylosing Spondylitis Targeting Osteocytes and Chondrocytes via Ameliorating Immune Dysfunctions. Cells 2023; 12:cells12070972. [PMID: 37048045 PMCID: PMC10093245 DOI: 10.3390/cells12070972] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 04/14/2023] Open
Abstract
Ankylosing spondylitis (AS) is clinically characterized by bone fusion that is induced by the pathological formation of extra bone. Unfortunately, the fundamental mechanism and related therapies remain unclear. The loss of SHP-2 (encoded by Ptpn11) in CD4-Cre;Ptpn11f/f mice resulted in the induction of AS-like pathological characteristics, including spontaneous cartilage and bone lesions, kyphosis, and arthritis. Hence, this mouse was utilized as an AS model in this study. As one of the basic physical fields, the magnetic field (MF) has been proven to be an effective treatment method for articular cartilage degeneration. In this study, the effects of a rotating magnetic field (RMF; 0.2 T, 4 Hz) on an AS-like mouse model were investigated. The RMF treatment (2 h/d, 0.2 T, 4 Hz) was performed on AS mice from two months after birth until the day before sampling. The murine specimens were subjected to transcriptomics, immunomics, and metabolomics analyses, combined with molecular and pathological experiments. The results demonstrated that the mitigation of inflammatory deterioration resulted in an increase in functional osteogenesis and a decrease in dysfunctional osteolysis due to the maintenance of bone homeostasis via the RANKL/RANK/OPG signaling pathway. Additionally, by regulating the ratio of CD4+ and CD8+ T-cells, RMF treatment rebalanced the immune microenvironment in skeletal tissue. It has been observed that RMF interventions have the potential to alleviate AS, including by decreasing pathogenicity and preventing disease initiation. Consequently, RMF, as a moderately physical therapeutic strategy, could be considered to alleviate the degradation of cartilage and bone tissue in AS and as a potential option to halt the progression of AS.
Collapse
Affiliation(s)
- Yu Han
- Magnetobiology Group, Department of Physiology, Shenzhen University Health Science Center, Xili Campus of Shenzhen University, Shenzhen 518055, China
| | - Hua Yang
- Magnetobiology Group, Department of Physiology, Shenzhen University Health Science Center, Xili Campus of Shenzhen University, Shenzhen 518055, China
| | - Zhongke Hua
- Magnetobiology Group, Department of Physiology, Shenzhen University Health Science Center, Xili Campus of Shenzhen University, Shenzhen 518055, China
| | - Shenglan Nie
- Magnetobiology Group, Department of Physiology, Shenzhen University Health Science Center, Xili Campus of Shenzhen University, Shenzhen 518055, China
| | - Shuling Xu
- Magnetobiology Group, Department of Physiology, Shenzhen University Health Science Center, Xili Campus of Shenzhen University, Shenzhen 518055, China
| | - Cai Zhou
- Magnetobiology Group, Department of Physiology, Shenzhen University Health Science Center, Xili Campus of Shenzhen University, Shenzhen 518055, China
| | - Fengyi Chen
- Magnetobiology Group, Department of Physiology, Shenzhen University Health Science Center, Xili Campus of Shenzhen University, Shenzhen 518055, China
| | - Mengqing Li
- Magnetobiology Group, Department of Physiology, Shenzhen University Health Science Center, Xili Campus of Shenzhen University, Shenzhen 518055, China
| | - Qinyao Yu
- Magnetobiology Group, Department of Physiology, Shenzhen University Health Science Center, Xili Campus of Shenzhen University, Shenzhen 518055, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Yunpeng Wei
- Magnetobiology Group, Department of Physiology, Shenzhen University Health Science Center, Xili Campus of Shenzhen University, Shenzhen 518055, China
| | - Xiaomei Wang
- Magnetobiology Group, Department of Physiology, Shenzhen University Health Science Center, Xili Campus of Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
14
|
Zhu L, Wang Z, Sun X, Yu J, Li T, Zhao H, Ji Y, Peng B, Du M. STAT3/Mitophagy Axis Coordinates Macrophage NLRP3 Inflammasome Activation and Inflammatory Bone Loss. J Bone Miner Res 2023; 38:335-353. [PMID: 36502520 DOI: 10.1002/jbmr.4756] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 11/06/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3), a cytokine-responsive transcription factor, is known to play a role in immunity and bone remodeling. However, whether and how STAT3 impacts macrophage NLR family pyrin domain containing 3 (NLRP3) inflammasome activation associated with inflammatory bone loss remains unknown. Here, STAT3 signaling is hyperactivated in macrophages in the context of both non-sterile and sterile inflammatory osteolysis, and this was highly correlated with the cleaved interleukin-1β (IL-1β) expression pattern. Strikingly, pharmacological inhibition of STAT3 markedly blocks macrophage NLRP3 inflammasome activation in vitro, thereby relieving inflammatory macrophage-amplified osteoclast formation and bone-resorptive activity. Mechanistically, STAT3 inhibition in macrophages triggers PTEN-induced kinase 1 (PINK1)-dependent mitophagy that eliminates dysfunctional mitochondria, reverses mitochondrial membrane potential collapse, and inhibits mitochondrial reactive oxygen species release, thus inactivating the NLRP3 inflammasome. In vivo, STAT3 inhibition effectively protects mice from both infection-induced periapical lesions and aseptic titanium particle-mediated calvarial bone erosion with potent induction of PINK1 and downregulation of inflammasome activation, macrophage infiltration, and osteoclast formation. This study reveals the regulatory role of the STAT3/mitophagy axis at the osteo-immune interface and highlights a potential therapeutic intervention to prevent inflammatory bone loss. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Lingxin Zhu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zijun Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiaoyue Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jingjing Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ting Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Huan Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yaoting Ji
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bin Peng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Minquan Du
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Bae S, Kim K, Kang K, Kim H, Lee M, Oh B, Kaneko K, Ma S, Choi JH, Kwak H, Lee EY, Park SH, Park-Min KH. RANKL-responsive epigenetic mechanism reprograms macrophages into bone-resorbing osteoclasts. Cell Mol Immunol 2023; 20:94-109. [PMID: 36513810 PMCID: PMC9794822 DOI: 10.1038/s41423-022-00959-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/03/2022] [Indexed: 12/15/2022] Open
Abstract
Monocyte/macrophage lineage cells are highly plastic and can differentiate into various cells under different environmental stimuli. Bone-resorbing osteoclasts are derived from the monocyte/macrophage lineage in response to receptor activator of NF-κB ligand (RANKL). However, the epigenetic signature contributing to the fate commitment of monocyte/macrophage lineage differentiation into human osteoclasts is largely unknown. In this study, we identified RANKL-responsive human osteoclast-specific superenhancers (SEs) and SE-associated enhancer RNAs (SE-eRNAs) by integrating data obtained from ChIP-seq, ATAC-seq, nuclear RNA-seq and PRO-seq analyses. RANKL induced the formation of 200 SEs, which are large clusters of enhancers, while suppressing 148 SEs in macrophages. RANKL-responsive SEs were strongly correlated with genes in the osteoclastogenic program and were selectively increased in human osteoclasts but marginally presented in osteoblasts, CD4+ T cells, and CD34+ cells. In addition to the major transcription factors identified in osteoclasts, we found that BATF binding motifs were highly enriched in RANKL-responsive SEs. The depletion of BATF1/3 inhibited RANKL-induced osteoclast differentiation. Furthermore, we found increased chromatin accessibility in SE regions, where RNA polymerase II was significantly recruited to induce the extragenic transcription of SE-eRNAs, in human osteoclasts. Knocking down SE-eRNAs in the vicinity of the NFATc1 gene diminished the expression of NFATc1, a major regulator of osteoclasts, and osteoclast differentiation. Inhibiting BET proteins suppressed the formation of some RANKL-responsive SEs and NFATc1-associated SEs, and the expression of SE-eRNA:NFATc1. Moreover, SE-eRNA:NFATc1 was highly expressed in the synovial macrophages of rheumatoid arthritis patients exhibiting high-osteoclastogenic potential. Our genome-wide analysis revealed RANKL-inducible SEs and SE-eRNAs as osteoclast-specific signatures, which may contribute to the development of osteoclast-specific therapeutic interventions.
Collapse
Affiliation(s)
- Seyeon Bae
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Kibyeong Kim
- Department of Biological Science, Ulsan National Institute of Science & Technology (UNIST), Ulsan, 44919, Republic of Korea
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Keunsoo Kang
- Department of Microbiology, Dankook University, Cheonan, 3116, Republic of Korea
| | - Haemin Kim
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Minjoon Lee
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Brian Oh
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Kaichi Kaneko
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Sungkook Ma
- Department of Biological Science, Ulsan National Institute of Science & Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jae Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Hojoong Kwak
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, USA
| | - Eun Young Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Sung Ho Park
- Department of Biological Science, Ulsan National Institute of Science & Technology (UNIST), Ulsan, 44919, Republic of Korea.
| | - Kyung-Hyun Park-Min
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
- BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, 10021, USA.
| |
Collapse
|
16
|
Yang G, Kang HC, Cho YY, Lee HS, Lee JY. Inflammasomes and their roles in arthritic disease pathogenesis. Front Mol Biosci 2022; 9:1027917. [PMID: 36387275 PMCID: PMC9650081 DOI: 10.3389/fmolb.2022.1027917] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/17/2022] [Indexed: 11/14/2023] Open
Abstract
The inflammasome is a molecular platform that is created in the cytosolic compartment to mediate the host immunological response to cellular injury and infection. Caspase-1 may be activated by the inflammasome, which leads to the generation of the inflammatory cytokines interleukin-1β (IL-1β) and IL-18 and the beginning of pyroptosis, which is a type of proinflammatory cell death. Scientists have identified a number of different inflammasomes in the last 2 decades. The NLRP3 inflammasome has been studied the most, and its activity may be triggered by a broad range of different inducers. However, activation of the NLRP3 inflammasome in a manner that is not properly controlled is also a factor in the etiology of many human illnesses. Accumulating evidence indicates that the NLRP3 inflammasome plays a significant role in the innate and adaptive immune systems and the development of various arthritic illnesses, such as rheumatoid arthritis, ankylosing spondylitis, and gout. The present review provides a concise summary of the biological properties of the NLRP3 inflammasome and presents the fundamental processes behind its activation and control. We discuss the role of the inflammasome in the pathogenesis of arthritic diseases, such as rheumatoid arthritis, ankylosing spondylitis, and gout, and the potential of newly developed therapies that specifically target the inflammasome or its products for the treatment of inflammatory diseases, with a particular emphasis on treatment and clinical application.
Collapse
Affiliation(s)
- Gabsik Yang
- Department of Pharmacology, College of Korean Medicine, Woosuk University, Jeonju, South Korea
| | - Han Chang Kang
- College of Pharmacy, The Catholic University of Korea, Seoul, South Korea
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Seoul, South Korea
| | - Hye Suk Lee
- College of Pharmacy, The Catholic University of Korea, Seoul, South Korea
| | - Joo Young Lee
- College of Pharmacy, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
17
|
Liu J, Zhang D, Cao Y, Zhang H, Li J, Xu J, Yu L, Ye S, Yang L. Screening of crosstalk and pyroptosis-related genes linking periodontitis and osteoporosis based on bioinformatics and machine learning. Front Immunol 2022; 13:955441. [PMID: 35990678 PMCID: PMC9389017 DOI: 10.3389/fimmu.2022.955441] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background and objective This study aimed to identify crosstalk genes between periodontitis (PD) and osteoporosis (OP) and potential relationships between crosstalk and pyroptosis-related genes. Methods PD and OP datasets were downloaded from the GEO database and were performed differential expression analysis to obtain DEGs. Overlapping DEGs got crosstalk genes linking PD and OP. Pyroptosis-related genes were obtained from literature reviews. Pearson coefficients were used to calculate crosstalk and pyroptosis-related gene correlations in the PD and OP datasets. Paired genes were obtained from the intersection of correlated genes in PD and OP. PINA and STRING databases were used to conduct the crosstalk-bridge-pyroptosis genes PPI network. The clusters in which crosstalk and pyroptosis-related genes were mainly concentrated were defined as key clusters. The key clusters' hub genes and the included paired genes were identified as key crosstalk-pyroptosis genes. Using ROC curve analysis and XGBoost screened key genes. PPI subnetwork, gene-biological process and gene-pathway networks were constructed based on key genes. In addition, immune infiltration was analyzed on the PD dataset using the CIBERSORT algorithm. Results A total of 69 crosstalk genes were obtained. 13 paired genes and hub genes TNF and EGFR in the key clusters (cluster2, cluster8) were identified as key crosstalk-pyroptosis genes. ROC and XGBoost showed that PRKCB, GSDMD, ARMCX3, and CASP3 were more accurate in predicting disease than other key crosstalk-pyroptosis genes while better classifying properties as a whole. KEGG analysis showed that PRKCB, GSDMD, ARMCX3, and CASP3 were involved in neutrophil extracellular trap formation and MAPK signaling pathway pathways. Immune infiltration results showed that all four key genes positively correlated with plasma cells and negatively correlated with T cells follicular helper, macrophages M2, and DCs. Conclusion This study shows a joint mechanism between PD and OP through crosstalk and pyroptosis-related genes. The key genes PRKCB, GSDMD, ARMCX3, and CASP3 are involved in the neutrophil extracellular trap formation and MAPK signaling pathway, affecting both diseases. These findings may point the way to future research.
Collapse
Affiliation(s)
- Jia Liu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Ding Zhang
- Department of Spine Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yu Cao
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Huichao Zhang
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Jianing Li
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Jingyu Xu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Ling Yu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Surong Ye
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Luyi Yang
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
18
|
Yin H, Liu N, Sigdel KR, Duan L. Role of NLRP3 Inflammasome in Rheumatoid Arthritis. Front Immunol 2022; 13:931690. [PMID: 35833125 PMCID: PMC9271572 DOI: 10.3389/fimmu.2022.931690] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by multi-articular, symmetrical and invasive arthritis resulting from immune system abnormalities involving T and B lymphocytes. Although significant progress has been made in the understanding of RA pathogenesis, the underlying mechanisms are not fully understood. Recent studies suggest that NLRP3 inflammasome, a regulator of inflammation, might play an important role in the development of RA. There have been increasing clinical and pre-clinical evidence showing the treatment of NLRP3/IL-1β in inflammatory diseases. To provide a foundation for the development of therapeutic strategies, we will briefly summarize the roles of NLRP3 inflammasome in RA and explore its potential clinical treatment.
Collapse
Affiliation(s)
- Hui Yin
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, China
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Na Liu
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, China
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Keshav Raj Sigdel
- Department of Internal Medicine, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Lihua Duan
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, China
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- *Correspondence: Lihua Duan,
| |
Collapse
|
19
|
Jiang Q, Wang X, Huang E, Wang Q, Wen C, Yang G, Lu L, Cui D. Inflammasome and Its Therapeutic Targeting in Rheumatoid Arthritis. Front Immunol 2022; 12:816839. [PMID: 35095918 PMCID: PMC8794704 DOI: 10.3389/fimmu.2021.816839] [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] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/21/2021] [Indexed: 12/30/2022] Open
Abstract
Inflammasome is a cytoplasmic multiprotein complex that facilitates the clearance of exogenous microorganisms or the recognition of endogenous danger signals, which is critically involved in innate inflammatory response. Excessive or abnormal activation of inflammasomes has been shown to contribute to the development of various diseases including autoimmune diseases, neurodegenerative changes, and cancers. Rheumatoid arthritis (RA) is a chronic and complex autoimmune disease, in which inflammasome activation plays a pivotal role in immune dysregulation and joint inflammation. This review summarizes recent findings on inflammasome activation and its effector mechanisms in the pathogenesis of RA and potential development of therapeutic targeting of inflammasome for the immunotherapy of RA.
Collapse
Affiliation(s)
- Qi Jiang
- Department of Blood Transfusion, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Xin Wang
- Department of Rheumatology and Immunology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Enyu Huang
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Chongqing International Institute for Immunology, Chongqing, China
| | - Qiao Wang
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chengping Wen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guocan Yang
- Department of Blood Transfusion, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Chongqing International Institute for Immunology, Chongqing, China
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
20
|
Wang C, Yang T, Xiao J, Xu C, Alippe Y, Sun K, Kanneganti TD, Monahan JB, Abu-Amer Y, Lieberman J, Mbalaviele G. NLRP3 inflammasome activation triggers gasdermin D-independent inflammation. Sci Immunol 2021; 6:eabj3859. [PMID: 34678046 DOI: 10.1126/sciimmunol.abj3859] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Chun Wang
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tong Yang
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jianqiu Xiao
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Canxin Xu
- Aclaris Therapeutics Inc., St. Louis, MO 63108, USA
| | - Yael Alippe
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kai Sun
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | | | | | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.,Shriners Hospital for Children, St. Louis, MO 63110, USA
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115 USA
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
21
|
Makkar R, Behl T, Bungau S, Kumar A, Arora S. Understanding the Role of Inflammasomes in Rheumatoid Arthritis. Inflammation 2021; 43:2033-2047. [PMID: 32712858 DOI: 10.1007/s10753-020-01301-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammasomes are the molecular pathways that activate upon conditions of infection or stress and trigger the activation and maturation of inflammatory cytokines. Immune reactions in conjugation with inflammatory processes play a pivotal role in developing innumerable diseases. An over reactive immune system fabricates many allergic and hypersensitive reactions in response to autoantibodies activated against modified self-epitopes and similar molecules. Rheumatoid arthritis (RA) is a complex autoimmune inflammatory disorder commencing with inflammation in small joints like hands, knees, and wrist eventually entrapping larger joints such as spine. The formation of autoantibodies called rheumatoid factor (RF) and citrullinated proteins against immunoglobulin G symbolizes autoimmune nature of the disease. The presence of autoantibodies embarks principal diagnostic hallmark of the disease. With the advancement of technology, the therapeutic approach is also advancing. A new era of molecules, namely inflammasomes, are activated upon infection or in response to stress and trigger the activation of various proinflammatory cytokines such interleukins which engage in the defense mechanism of the innate immunity. Robust linking among the activity of dysregulated inflammasomes and the heritable acquired inflammatory diseases and disorders emphasizes the significance of this pathway in altering the immune responses. The current review highlights the functioning of inflammasomes and their possible role in disease dysregulation.
Collapse
Affiliation(s)
- Rashita Makkar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| |
Collapse
|
22
|
Burdette BE, Esparza AN, Zhu H, Wang S. Gasdermin D in pyroptosis. Acta Pharm Sin B 2021; 11:2768-2782. [PMID: 34589396 PMCID: PMC8463274 DOI: 10.1016/j.apsb.2021.02.006] [Citation(s) in RCA: 328] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023] Open
Abstract
Pyroptosis is the process of inflammatory cell death. The primary function of pyroptosis is to induce strong inflammatory responses that defend the host against microbe infection. Excessive pyroptosis, however, leads to several inflammatory diseases, including sepsis and autoimmune disorders. Pyroptosis can be canonical or noncanonical. Upon microbe infection, the canonical pathway responds to pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), while the noncanonical pathway responds to intracellular lipopolysaccharides (LPS) of Gram-negative bacteria. The last step of pyroptosis requires the cleavage of gasdermin D (GsdmD) at D275 (numbering after human GSDMD) into N- and C-termini by caspase 1 in the canonical pathway and caspase 4/5/11 (caspase 4/5 in humans, caspase 11 in mice) in the noncanonical pathway. Upon cleavage, the N-terminus of GsdmD (GsdmD-N) forms a transmembrane pore that releases cytokines such as IL-1β and IL-18 and disturbs the regulation of ions and water, eventually resulting in strong inflammation and cell death. Since GsdmD is the effector of pyroptosis, promising inhibitors of GsdmD have been developed for inflammatory diseases. This review will focus on the roles of GsdmD during pyroptosis and in diseases.
Collapse
Key Words
- 7DG, 7-desacetoxy-6,7-dehydrogedunin
- ADRA2B, α-2B adrenergic receptor
- AIM, absent in melanoma
- ASC, associated speck-like protein
- Ac-FLTD-CMK, acetyl-FLTD-chloromethylketone
- BMDM, bone marrow-derived macrophages
- CARD, caspase activation
- CD, Crohn’s disease
- CTM, Chinese traditional medicine
- CTSG, cathepsin G
- Caspase
- DAMP, damage-associated molecular pattern
- DFNA5, deafness autosomal dominant 5
- DFNB59, deafness autosomal recessive type 59
- DKD, diabetic kidney disease
- DMF, dimethyl fumarate
- Damage-associated molecular patterns (DAMPs)
- ELANE, neutrophil expressed elastase
- ESCRT, endosomal sorting complexes required for transport
- FADD, FAS-associated death domain
- FDA, U.S. Food and Drug Administration
- FIIND, function to find domain
- FMF, familial Mediterranean fever
- GI, gastrointestinal
- GPX, glutathione peroxidase
- Gasdermin
- GsdmA/B/C/D/E, gasdermin A/B/C/D/E
- HAMP, homeostasis altering molecular pattern
- HIN, hematopoietic expression, interferon-inducible nature, and nuclear localization
- HIV, human immunodeficiency virus
- HMGB1, high mobility group protein B1
- IBD, inflammatory bowel disease
- IFN, interferon
- ITPR1, inositol 1,4,5-trisphosphate receptor type 1
- Inflammasome
- Inflammation
- LPS, lipopolysaccharide
- LRR, leucine-rich repeat
- MAP3K7, mitogen-activated protein kinase kinase kinase 7
- MCC950, N-[[(1,2,3,5,6,7-hexahydro-s-indacen-4-yl)amino]carbonyl]-4-(1-hydroxy-1-methylethyl)-2-furansulfonamide
- NAIP, NLR family apoptosis inhibitory protein
- NBD, nucleotide-binding domain
- NEK7, NIMA-related kinase 7
- NET, neutrophil extracellular trap
- NIK, NF-κB inducing kinase
- NLR, NOD-like receptor
- NLRP, NLR family pyrin domain containing
- NSAID, non-steroidal anti-inflammatory drug
- NSCLC, non-small cell lung cancer
- NSP, neutrophil specific serine protease
- PAMP, pathogen-associated molecular pattern
- PKA, protein kinase A
- PKN1/2, protein kinase1/2
- PKR, protein kinase-R
- PRR, pattern recognition receptors
- PYD, pyrin domain
- Pathogen-associated molecular patterns (PAMPs)
- Pyroptosis
- ROS, reactive oxygen species
- STING, stimulator of interferon genes
- Sepsis
- TLR, Toll-like receptor
- UC, ulcerative colitis
- cAMP, cyclic adenosine monophosphate
- cGAS, cyclic GMP–AMP synthase
- mtDNA, mitochondrial DNA
Collapse
Affiliation(s)
- Brandon E. Burdette
- Biology Department, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| | - Ashley N. Esparza
- Biology Department, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| | - Hua Zhu
- Department of Surgery, the Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Shanzhi Wang
- Biology Department, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| |
Collapse
|
23
|
Hu Y, Wang Y, Chen T, Hao Z, Cai L, Li J. Exosome: Function and Application in Inflammatory Bone Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6324912. [PMID: 34504641 PMCID: PMC8423581 DOI: 10.1155/2021/6324912] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/18/2021] [Indexed: 12/26/2022]
Abstract
In the skeletal system, inflammation is closely associated with many skeletal disorders, including periprosthetic osteolysis (bone loss around orthopedic implants), osteoporosis, and rheumatoid arthritis. These diseases, referred to as inflammatory bone diseases, are caused by various oxidative stress factors in the body, resulting in long-term chronic inflammatory processes and eventually causing disturbances in bone metabolism, increased osteoclast activity, and decreased osteoblast activity, thereby leading to osteolysis. Inflammatory bone diseases caused by nonbacterial factors include inflammation- and bone resorption-related processes. A growing number of studies show that exosomes play an essential role in developing and progressing inflammatory bone diseases. Mechanistically, exosomes are involved in the onset and progression of inflammatory bone disease and promote inflammatory osteolysis, but specific types of exosomes are also involved in inhibiting this process. Exosomal regulation of the NF-κB signaling pathway affects macrophage polarization and regulates inflammatory responses. The inflammatory response further causes alterations in cytokine and exosome secretion. These signals regulate osteoclast differentiation through the receptor activator of the nuclear factor-kappaB ligand pathway and affect osteoblast activity through the Wnt pathway and the transcription factor Runx2, thereby influencing bone metabolism. Overall, enhanced bone resorption dominates the overall mechanism, and over time, this imbalance leads to chronic osteolysis. Understanding the role of exosomes may provide new perspectives on their influence on bone metabolism in inflammatory bone diseases. At the same time, exosomes have a promising future in diagnosing and treating inflammatory bone disease due to their unique properties.
Collapse
Affiliation(s)
- Yingkun Hu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lin Cai
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
24
|
Alippe Y, Kress D, Ricci B, Sun K, Yang T, Wang C, Xiao J, Abu-Amer Y, Mbalaviele G. Actions of the NLRP3 and NLRC4 inflammasomes overlap in bone resorption. FASEB J 2021; 35:e21837. [PMID: 34383985 DOI: 10.1096/fj.202100767rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022]
Abstract
Overwhelming evidence indicates that excessive stimulation of innate immune receptors of the NOD-like receptor (NLR) family causes significant damage to multiple tissues, yet the role of these proteins in bone metabolism is not well known. Here, we studied the interaction between the NLRP3 and NLRC4 inflammasomes in bone homeostasis and disease. We found that loss of NLRP3 or NLRC4 inflammasome attenuated osteoclast differentiation in vitro. At the tissue level, lack of NLRP3, or NLRC4 to a lesser extent, resulted in higher baseline bone mass compared to wild-type (WT) mice, and conferred protection against LPS-induced inflammatory osteolysis. Bone mass accrual in mutant mice correlated with lower serum IL-1β levels in vivo. Unexpectedly, the phenotype of Nlrp3-deficient mice was reversed upon loss of NLRC4 as bone mass was comparable between WT mice and Nlrp3;Nlrc4 knockout mice. Thus, although bone homeostasis is perturbed to various degrees by the lack of NLRP3 or NLRC4, this tissue appears to function normally upon compound loss of the inflammasomes assembled by these receptors.
Collapse
Affiliation(s)
- Yael Alippe
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Dustin Kress
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Biancamaria Ricci
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Kai Sun
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, USA.,Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Tong Yang
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, USA.,Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Chun Wang
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - Jianqiu Xiao
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 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
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
25
|
Szekanecz Z, McInnes IB, Schett G, Szamosi S, Benkő S, Szűcs G. Autoinflammation and autoimmunity across rheumatic and musculoskeletal diseases. Nat Rev Rheumatol 2021; 17:585-595. [PMID: 34341562 DOI: 10.1038/s41584-021-00652-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 12/16/2022]
Abstract
Most rheumatic and musculoskeletal diseases (RMDs) can be placed along a spectrum of disorders, with autoinflammatory diseases (including monogenic systemic autoinflammatory diseases) and autoimmune diseases (such as systemic lupus erythematosus and antiphospholipid syndrome) representing the two ends of this spectrum. However, although most autoinflammatory diseases are characterized by the activation of innate immunity and inflammasomes and classical autoimmunity typically involves adaptive immune responses, there is some overlap in the features of autoimmunity and autoinflammation in RMDs. Indeed, some 'mixed-pattern' diseases such as spondyloarthritis and some forms of rheumatoid arthritis can also be delineated. A better understanding of the pathogenic pathways of autoinflammation and autoimmunity in RMDs, as well as the preferential cytokine patterns observed in these diseases, could help us to design targeted treatment strategies.
Collapse
Affiliation(s)
- Zoltán Szekanecz
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum fur Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Szilvia Szamosi
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilvia Benkő
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gabriella Szűcs
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
26
|
The Role of Melatonin on NLRP3 Inflammasome Activation in Diseases. Antioxidants (Basel) 2021; 10:antiox10071020. [PMID: 34202842 PMCID: PMC8300798 DOI: 10.3390/antiox10071020] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/22/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
NLRP3 inflammasome is a part of the innate immune system and responsible for the rapid identification and eradication of pathogenic microbes, metabolic stress products, reactive oxygen species, and other exogenous agents. NLRP3 inflammasome is overactivated in several neurodegenerative, cardiac, pulmonary, and metabolic diseases. Therefore, suppression of inflammasome activation is of utmost clinical importance. Melatonin is a ubiquitous hormone mainly produced in the pineal gland with circadian rhythm regulatory, antioxidant, and immunomodulatory functions. Melatonin is a natural product and safer than most chemicals to use for medicinal purposes. Many in vitro and in vivo studies have proved that melatonin alleviates NLRP3 inflammasome activity via various intracellular signaling pathways. In this review, the effect of melatonin on the NLRP3 inflammasome in the context of diseases will be discussed.
Collapse
|
27
|
Li Y, Ling J, Jiang Q. Inflammasomes in Alveolar Bone Loss. Front Immunol 2021; 12:691013. [PMID: 34177950 PMCID: PMC8221428 DOI: 10.3389/fimmu.2021.691013] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022] Open
Abstract
Bone remodeling is tightly controlled by osteoclast-mediated bone resorption and osteoblast-mediated bone formation. Fine tuning of the osteoclast-osteoblast balance results in strict synchronization of bone resorption and formation, which maintains structural integrity and bone tissue homeostasis; in contrast, dysregulated bone remodeling may cause pathological osteolysis, in which inflammation plays a vital role in promoting bone destruction. The alveolar bone presents high turnover rate, complex associations with the tooth and periodontium, and susceptibility to oral pathogenic insults and mechanical stress, which enhance its complexity in host defense and bone remodeling. Alveolar bone loss is also involved in systemic bone destruction and is affected by medication or systemic pathological factors. Therefore, it is essential to investigate the osteoimmunological mechanisms involved in the dysregulation of alveolar bone remodeling. The inflammasome is a supramolecular protein complex assembled in response to pattern recognition receptors and damage-associated molecular patterns, leading to the maturation and secretion of pro-inflammatory cytokines and activation of inflammatory responses. Pyroptosis downstream of inflammasome activation also facilitates the clearance of intracellular pathogens and irritants. However, inadequate or excessive activity of the inflammasome may allow for persistent infection and infection spreading or uncontrolled destruction of the alveolar bone, as commonly observed in periodontitis, periapical periodontitis, peri-implantitis, orthodontic tooth movement, medication-related osteonecrosis of the jaw, nonsterile or sterile osteomyelitis of the jaw, and osteoporosis. In this review, we present a framework for understanding the role and mechanism of canonical and noncanonical inflammasomes in the pathogenesis and development of etiologically diverse diseases associated with alveolar bone loss. Inappropriate inflammasome activation may drive alveolar osteolysis by regulating cellular players, including osteoclasts, osteoblasts, osteocytes, periodontal ligament cells, macrophages, monocytes, neutrophils, and adaptive immune cells, such as T helper 17 cells, causing increased osteoclast activity, decreased osteoblast activity, and enhanced periodontium inflammation by creating a pro-inflammatory milieu in a context- and cell type-dependent manner. We also discuss promising therapeutic strategies targeting inappropriate inflammasome activity in the treatment of alveolar bone loss. Novel strategies for inhibiting inflammasome signaling may facilitate the development of versatile drugs that carefully balance the beneficial contributions of inflammasomes to host defense.
Collapse
Affiliation(s)
- Yang Li
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Junqi Ling
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China.,Guangdong Province Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qianzhou Jiang
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| |
Collapse
|
28
|
Chao-Yang G, Peng C, Hai-Hong Z. Roles of NLRP3 inflammasome in intervertebral disc degeneration. Osteoarthritis Cartilage 2021; 29:793-801. [PMID: 33609693 DOI: 10.1016/j.joca.2021.02.204] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/21/2020] [Accepted: 02/08/2021] [Indexed: 02/02/2023]
Abstract
Intervertebral disc degeneration (IVDD) is one of the leading causes of low back pain and one of the most common health problems in the world. The nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing-3 (NLRP3) inflammasome, as a pattern recognition receptor, has been shown to be associated with the pathological processes of many diseases in recent years. With the exploration of the mechanism of IVDD, recent studies have shown that activation of the NLRP3 inflammasome is associated with intervertebral disc (IVD) inflammation, pyroptosis, extracellular matrix degradation and apoptosis of IVD cells. In this review, we summarize the structural characteristics of NLRP3 inflammasome and the activation signalling mechanisms. We also describe the role of the NLRP3 inflammasome in the pathological process of IVDD and the application of the targeting the NLRP3 inflammasome in IVDD treatment.
Collapse
Affiliation(s)
- G Chao-Yang
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, PR China
| | - C Peng
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, 730000, PR China
| | - Z Hai-Hong
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, PR China.
| |
Collapse
|
29
|
Cao Y, Li L, Fan Z. The role and mechanisms of polycomb repressive complex 2 on the regulation of osteogenic and neurogenic differentiation of stem cells. Cell Prolif 2021; 54:e13032. [PMID: 33759287 PMCID: PMC8088470 DOI: 10.1111/cpr.13032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/25/2021] [Accepted: 03/11/2021] [Indexed: 12/25/2022] Open
Abstract
The stem cells differentiate into osteoblasts or neurocytes is the key process for treatment of bone‐ or neural tissue‐related diseases which is caused by ageing, fracture, injury, inflammation, etc Polycomb group complexes (PcGs), especially the polycomb repressive complex 2 (PRC2), act as pivotal epigenetic regulators by modifying key developmental regulatory genes during stem cells differentiation. In this review, we summarize the core subunits, the variants and the potential functions of PRC2. We also highlight the underlying mechanisms of PRC2 associated with the osteogenic and neurogenic differentiation of stem cells, including its interaction with non‐coding RNAs, histone acetyltransferases, histone demethylase, DNA methyltransferase and polycomb repressive complex 1. This review provided a substantial information of epigenetic regulation mediated by PRC2 which leads to the osteogenic and neurogenic differentiation of stem cells.
Collapse
Affiliation(s)
- Yangyang Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Le Li
- Tsinghua University Hospital, Stomatological Disease Prevention and Control Center, Tsinghua University, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
30
|
De Haan P, Van Diemen FR, Toscano MG. Viral gene delivery vectors: the next generation medicines for immune-related diseases. Hum Vaccin Immunother 2021; 17:14-21. [PMID: 32412865 PMCID: PMC7872028 DOI: 10.1080/21645515.2020.1757989] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
Viruses have evolved to efficiently express their genes in host cells, which makes them ideally suited as gene delivery vectors for gene and immunotherapies. Replication competent (RC) viral vectors encoding foreign or self-proteins induce strong T-cell responses that can be used for the development of effective cancer treatments. Replication-defective (RD) viral vectors encoding self-proteins are non-immunogenic when introduced in a host naïve for the cognate virus. RD viral vectors can be used to develop gene replacement therapies for genetic disorders and tolerization therapies for autoimmune diseases and allergies. Degenerative/inflammatory diseases are associated with chronic inflammation and immune responses that damage the tissues involved. These diseases therefore strongly resemble autoimmune diseases. This review deals with the use of RC and RD viral vectors for unraveling the pathogenesis of immune-related diseases and their application to the development of the next generation prophylactics and therapeutics for todays' major diseases.
Collapse
Affiliation(s)
- Peter De Haan
- Department of R&D, Amarna Therapeutics B.V, Leiden, The Netherlands
| | | | | |
Collapse
|
31
|
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: 3.8] [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
|
32
|
Chen YJ, Wang X, Zhang HY, Du YN, Jin XJ, Zhang ZF. Inverse association between bone mineral density and fibrinogen in menopausal women. Climacteric 2020; 24:146-150. [PMID: 32602367 DOI: 10.1080/13697137.2020.1776249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Inflammatory diseases are risk factors for osteoporosis. We aimed to explore whether fibrinogen, which is linked to chronic inflammation, is associated with bone mineral density (BMD) in menopausal women. METHODS In this cross-sectional study, we analyzed 339 menopausal women from Zhejiang Province between January 2016 and October 2019. Linear regression analysis was performed to assess the relationship between fibrinogen and BMD. RESULTS Significant inverse association was observed between the serum fibrinogen level and BMD in menopausal women. The mean BMD in each quartile of fibrinogen level was 0.901, 0.897, 0.892, and 0.855 g/cm2, respectively (p = 0.027). After adjusting for age, body mass index, metabolic profiles, blood inflammatory factors, and serum levels of estradiol, calcium, phosphorus, and alkaline phosphatase, fibrinogen levels remained significantly associated with BMD (regression coefficients for quartiles 1-3 vs. quartile 4 were 0.046, 0.027, and 0.036, respectively; p for trend <0.05). CONCLUSIONS Higher fibrinogen levels were associated with lower BMD in menopausal women, which was independent of age, body mass index, estradiol, and other factors. Therefore, serum fibrinogen can be used as a new predictor of reduced BMD in menopausal women.
Collapse
Affiliation(s)
- Y J Chen
- Department of fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - X Wang
- Nanjing Medical University, Nanjing, China
| | - H Y Zhang
- Department of Obstetrics and Gynecology, Hangzhou Women's Hospital, Hangzhou, China
| | - Y N Du
- Nanjing Medical University, Nanjing, China
| | - X J Jin
- Department of Obstetrics and Gynecology, Hangzhou Women's Hospital, Hangzhou, China
| | - Z F Zhang
- Department of fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.,Nanjing Medical University, Nanjing, China.,Department of Obstetrics and Gynecology, Hangzhou Women's Hospital, Hangzhou, China
| |
Collapse
|
33
|
Goodman SB, Gallo J. Periprosthetic Osteolysis: Mechanisms, Prevention and Treatment. J Clin Med 2019; 8:E2091. [PMID: 31805704 PMCID: PMC6947309 DOI: 10.3390/jcm8122091] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 02/06/2023] Open
Abstract
Clinical studies, as well as in vitro and in vivo experiments have demonstrated that byproducts from joint replacements induce an inflammatory reaction that can result in periprosthetic osteolysis (PPOL) and aseptic loosening (AL). Particle-stimulated macrophages and other cells release cytokines, chemokines, and other pro-inflammatory substances that perpetuate chronic inflammation, induce osteoclastic bone resorption and suppress bone formation. Differentiation, maturation, activation, and survival of osteoclasts at the bone-implant interface are under the control of the receptor activator of nuclear factor kappa-Β ligand (RANKL)-dependent pathways, and the transcription factors like nuclear factor κB (NF-κB) and activator protein-1 (AP-1). Mechanical factors such as prosthetic micromotion and oscillations in fluid pressures also contribute to PPOL. The treatment for progressive PPOL is only surgical. In order to mitigate ongoing loss of host bone, a number of non-operative approaches have been proposed. However, except for the use of bisphosphonates in selected cases, none are evidence based. To date, the most successful and effective approach to preventing PPOL is usage of wear-resistant bearing couples in combination with advanced implant designs, reducing the load of metallic and polymer particles. These innovations have significantly decreased the revision rate due to AL and PPOL in the last decade.
Collapse
Affiliation(s)
- Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, 450 Broadway St. M/C 6342, Redwood City, CA 94063, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Jiri Gallo
- Department of Orthopaedics, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, I. P. Pavlova 6, 779 00 Olomouc, Czech Republic;
| |
Collapse
|
34
|
Wang C, Mbalaviele G. Role of APD-Ribosylation in Bone Health and Disease. Cells 2019; 8:cells8101201. [PMID: 31590342 PMCID: PMC6829334 DOI: 10.3390/cells8101201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022] Open
Abstract
The transfer of adenosine diphosphate (ADP)-ribose unit(s) from nicotinamide adenine dinucleotide (NAD+) to acceptor proteins is known as ADP-ribosylation. This post-translational modification (PTM) unavoidably alters protein functions and signaling networks, thereby impacting cell behaviors and tissue outcomes. As a ubiquitous mechanism, ADP-ribosylation affects multiple tissues, including bones, as abnormal ADP-ribosylation compromises bone development and remodeling. In this review, we describe the effects of ADP-ribosylation in bone development and maintenance, and highlight the underlying mechanisms.
Collapse
Affiliation(s)
- Chun Wang
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
35
|
Affiliation(s)
- Mary C Nakamura
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA.
- Arthritis/Immunology Section, San Francisco Veterans Administration Health Care System, 4150 Clement Street, 111R, San Francisco, CA, 94121, USA.
| |
Collapse
|