1
|
Ding X, Yang J, Wei Y, Wang M, Peng Z, He R, Li X, Zhao D, Leng X, Dong H. The Nexus Between Traditional Chinese Medicine and Immunoporosis: Implications in the Treatment and Management of Osteoporosis. Phytother Res 2025; 39:1826-1846. [PMID: 39625224 DOI: 10.1002/ptr.8397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/21/2024] [Accepted: 11/06/2024] [Indexed: 01/06/2025]
Abstract
Osteoporosis (OP) is a globally prevalent bone disease characterized by reduced bone mass and heightened fracture risk, posing a significant health and economic challenge to aging societies worldwide. Osteoimmunology-an emerging field of study-investigates the intricate relationship between the skeletal and the immune systems, providing insights into the immune system's impact on bone health and disease progression. Recent research has demonstrated the essential roles played by various immune cells (T cells, B cells, macrophages, dendritic cells, mast cells, granulocytes, and innate lymphoid cells) in regulating bone metabolism, homeostasis, formation, and remodeling through interactions with osteoclasts (OC) and osteoblasts (OB). These findings underscore that osteoimmunology provides an essential theoretical framework for understanding the pathogenesis of various skeletal disorders, including OP. Traditional Chinese medicine (TCM) and its active ingredients have significant clinical value in OP treatment. Unfortunately, despite their striking multieffect pathways in the pharmacological field, current research has not yet summarized them in a comprehensive and detailed manner with respect to their interventional roles in immune bone diseases, especially OP. Consequently, this review addresses recent studies on the mechanisms by which immune cells and their communication molecules contribute to OP development. Additionally, it explores the potential therapeutic benefits of TCM and its active components in treating OP from the perspective of osteoimmunology. The objective is to provide a comprehensive framework that enhances the understanding of the therapeutic mechanisms of TCM in treating immune-related bone diseases and to facilitate the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Xiaolei Ding
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jie Yang
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yuchi Wei
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Mingyue Wang
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Zeyu Peng
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Rong He
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiangyan Li
- Northeast Asia Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Daqing Zhao
- Northeast Asia Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiangyang Leng
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Haisi Dong
- Northeast Asia Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
2
|
Liu Y, Yang Y, Li Y, Ding W, Yang X. Association between lipid accumulation products and mortality outcomes in patients with osteoporosis and osteopenia. Exp Gerontol 2025; 201:112705. [PMID: 39914581 DOI: 10.1016/j.exger.2025.112705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND Osteoporosis (OP) and osteopenia are metabolic bone disorders associated with increased fragility and fracture risk. While lipid accumulation products (LAP) are emerging as potential markers of metabolic health, their prognostic significance in patients with OP or osteopenia remains unclear. The objective of this study is to elucidate the relationship between lipid accumulation products (LAP) and all-cause as well as cardiovascular mortality in individuals diagnosed with either condition. METHODS Data from the 2007-2018 National Health and Nutrition Examination Survey (NHANES) were retrospectively analyzed. Kaplan-Meier survival curves, multivariable Cox proportional hazards regression, and restricted cubic spline plots were used to evaluate the association between LAP and mortality outcomes in patients with OP or osteopenia. Subgroup and threshold analyses were also conducted. RESULTS This study included 4959 patients diagnosed with OP or osteopenia, followed over a comprehensive duration of 12 years, during which 800 instances of all-cause mortality and 194 deaths attributed to cardiovascular diseases were documented. A linear negative correlation was identified between LAP and both all-cause and cardiovascular mortality among patients with OP or osteopenia. Notably, at an LAP level of 3.69, the risk ratio reached 1, indicating a transition in mortality risk from high to low. Subgroup analyses revealed a more pronounced association between LAP and mortality. CONCLUSION Our study revealed a significant linear negative correlation between the lipid accumulation product (LAP) and both all-cause and cardiovascular mortality in patients diagnosed with osteoporosis (OP) and osteopenia.
Collapse
Affiliation(s)
- Yazhou Liu
- Department of Orthopedics, Dalian Medical University, Dalian, China; Department of Orthopedics, Dandong Central Hospital, Dalian Medical University, Dandong, China
| | - Ying Yang
- Department of Gynecology, Dalian Medical University, Dalian, China
| | - Yuhao Li
- Department of Orthopedics, Dandong Central Hospital, China Medical University, Dandong, China
| | - Wenbo Ding
- Department of Orthopedics, Dandong Central Hospital, China Medical University, Dandong, China
| | - Xiaodong Yang
- Department of Orthopedics, Dandong Central Hospital, Dalian Medical University, Dandong, China.
| |
Collapse
|
3
|
Ben Amara H, Martinez DC, Iskhakova K, Emanuelsson L, Norlindh B, Johansson Loo A, Wieland DCF, Zeller-Plumhoff B, Willumeit-Römer R, Plocinski T, Swieszkowski W, Shah FA, Palmquist A, Omar O, Thomsen P. Multifaceted bone response to immunomodulatory magnesium implants: Osteopromotion at the interface and adipogenesis in the bone marrow. Biomaterials 2025; 314:122779. [PMID: 39305536 DOI: 10.1016/j.biomaterials.2024.122779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 08/05/2024] [Accepted: 08/27/2024] [Indexed: 11/10/2024]
Abstract
Orthopedic implants made of biodegradable magnesium (Mg) provide an alternative to nondegradable implants for fracture repair. Widely reported to be pro-osteogenic, Mg implants are also believed to be anti-inflammatory and anti-osteoclastic, but this is difficult to reconcile with the early clinical inflammation observed around these implants. Here, by surveying implant healing in a rat bone model, we determined the cellular responses and structural assembly of bone correlated with the surface changes of Mg implants inherent in degradation. We show that, compared to titanium, both high-purity (99.998 %) and clinical-grade, rare earth-alloyed (MgYREZr) Mg implants create an initial, transient proinflammatory environment that facilitates inducible nitric oxide synthase-mediated macrophage polarization, osteoclastogenesis, and neoangiogenesis programs. While this immunomodulation subsequently reinforces reparative osteogenesis at the surface of both Mg implants, the faster degradation of high-purity Mg implants, but not MgYREZr implants, elicits a compositional alteration in the interfacial bone and a previously unknown proadipogenic response with persistent low-grade inflammation in the surrounding bone marrow. Beyond the need for rigorous tailoring of Mg implants, these data highlight the need to closely monitor osseointegration not only at the immediate implant surface but also in the peri-implant bone and adjacent bone marrow.
Collapse
Affiliation(s)
- Heithem Ben Amara
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Diana C Martinez
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Poland
| | - Kamila Iskhakova
- Institute of Metallic Biomaterials, Helmholtz-Zentrum Hereon, Geesthacht, Germany
| | - Lena Emanuelsson
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Birgitta Norlindh
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Anna Johansson Loo
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - D C Florian Wieland
- Institute of Metallic Biomaterials, Helmholtz-Zentrum Hereon, Geesthacht, Germany
| | | | | | - Tomasz Plocinski
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Poland
| | - Wojciech Swieszkowski
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Poland
| | - Furqan A Shah
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Anders Palmquist
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Omar Omar
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Peter Thomsen
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
4
|
Wang H, Li Y, Li H, Yan X, Jiang Z, Feng L, Hu W, Fan Y, Lin S, Li G. T cell related osteoimmunology in fracture healing: Potential targets for augmenting bone regeneration. J Orthop Translat 2025; 51:82-93. [PMID: 39991456 PMCID: PMC11847249 DOI: 10.1016/j.jot.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/15/2024] [Accepted: 12/01/2024] [Indexed: 02/25/2025] Open
Abstract
UNLABELLED Last decade has witnessed increasing evidence which highlights the roles of immune cells in bone regeneration. Numerous immune cell types, including macrophages, T cells, and neutrophils are involved in fracture healing by orchestrating a series of events that modulate bone formation and remodeling. In this review, the role of T cell immunity in fracture healing has been summarized, and the modulatory effects of T cell immunity in inflammation, bone formation and remodeling have been highlighted. The review also summarizes the specific roles of different T cell subsets, including CD4+ T cells, CD8+ T cells, regulatory T cells, T helper 17 cells, and γδ T cells in modulating fracture healing. The current therapeutics targeting T cell immunity to enhance fracture healing have also been reviewed, aiming to provide insights from a translational standpoint. Overall, this work discusses recent advances and challenges in the interdisciplinary research field of T cell related osteoimmunology and its implications in fracture healing. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE Delayed unions or non-unions of bone fractures remain a challenge in clinical practice. Developing a deep understanding of the roles of immune cells, including T cells, in fracture healing will facilitate the advancement of novel therapeutics of fracture nonunion. This review summarizes the current understanding of different T cell subsets involved in various phases of fracture healing, providing insights for targeting T cells as an alternative strategy to enhance bone regeneration.
Collapse
Affiliation(s)
- Haixing Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yashi Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haoxin Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xu Yan
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhaowei Jiang
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lu Feng
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China
| | - Wenhui Hu
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Yinuo Fan
- The Third Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Gang Li
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
Yao Y, Cai X, Chen Y, Zhang M, Zheng C. Estrogen deficiency-mediated osteoimmunity in postmenopausal osteoporosis. Med Res Rev 2025; 45:561-575. [PMID: 39234932 DOI: 10.1002/med.22081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/03/2023] [Accepted: 08/25/2024] [Indexed: 09/06/2024]
Abstract
Postmenopausal osteoporosis (PMO) is a common disease associated with aging, and estrogen deficiency is considered to be the main cause of PMO. Recently, however, osteoimmunology has been revealed to be closely related to PMO. On the one hand, estrogen deficiency directly affects the activity of bone cells (osteoblasts, osteoclasts, osteocytes). On the other hand, estrogen deficiency-mediated osteoimmunity also plays a crucial role in bone loss in PMO. In this review, we systematically describe the progress of the mechanisms of bone loss in PMO, estrogen deficiency-mediated osteoimmunity, the differences between PMO patients and postmenopausal populations without osteoporosis, and estrogen deficiency-mediated immune cells (T cells, B cells, macrophages, neutrophils, dendritic cells, and mast cells) activity. The comprehensive summary of this paper provides a clear knowledge context for future research on the mechanism of PMO bone loss.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Xiaoyu Cai
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
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
|
7
|
King JS, Wan M, Kim A, Novak S, Prabhu S, Kalajzic I, Delany AM, Sanjay A. Effects of Aging on the Immune and Periosteal Response to Fracture in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622348. [PMID: 39574733 PMCID: PMC11580938 DOI: 10.1101/2024.11.06.622348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Aging predisposes individuals to reduced bone mass and fragility fractures, which are costly and linked to high mortality. Understanding how aging affects fracture healing is essential for developing therapies to enhance bone regeneration in older adults. During the inflammatory phase of fracture healing, immune cells are recruited to the injury site as periosteal skeletal stem/progenitor cells (pSSPCs) rapidly proliferate and differentiate into osteochondral lineages, allowing for fibrocartilaginous callus formation and complete bone healing. Irrespective of age, how periosteal mesenchymal and immune cells interact during early fracture healing is incompletely understood, limiting our ability to potentially modulate these processes. To address this, we directly analyzed, in parallel, at a single-cell level, isolated murine CD45(+) and CD45(-) periosteal cells dissected from intact and fractured bones, collected three days after injury. Through comprehensive analysis, corroborated by bulk RNA-sequencing, flow cytometry, and histology, we found aging decreases pSSPCs proliferative, marked by a reduced expression of genes required for callus formation and an increased senescence signature. We found that the chemokine Cxcl9 was highly upregulated in aged intact Prrx1+ pSSPCs, predicted to interact with other pSSPCs directly, and associated with increased recruitment of CD8+ T cells at the fracture site three days after injury. Cell-to-cell communication analysis provided insight into the complexity of interactions among the many cell types regulating fracture healing and the impact of aging on these processes. Together, these results provide insight into age-induced alterations in fracture healing, informing the development of improved therapeutic approaches for fragility fractures.
Collapse
|
8
|
Maouche A, Boumediene K, Baugé C. Bioactive Compounds in Osteoarthritis: Molecular Mechanisms and Therapeutic Roles. Int J Mol Sci 2024; 25:11656. [PMID: 39519204 PMCID: PMC11546619 DOI: 10.3390/ijms252111656] [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: 10/08/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Osteoarthritis (OA) is the most common and debilitating form of arthritis. Current therapies focus on pain relief and efforts to slow disease progression through a combination of drug and non-drug treatments. Bioactive compounds derived from plants show significant promise due to their anti-inflammatory, antioxidant, and tissue-protective properties. These natural compounds can help regulate the inflammatory processes and metabolic pathways involved in OA, thereby alleviating symptoms and potentially slowing disease progression. Investigating the efficacy of these natural agents in treating osteoarthritis addresses a growing demand for natural health solutions and creates new opportunities for managing this increasingly prevalent age-related condition. The aim of this review is to provide an overview of the use of some bioactive compounds from plants in modulating the progression of osteoarthritis and alleviating associated pain.
Collapse
Affiliation(s)
| | | | - Catherine Baugé
- UR7451 BIOCONNECT, Université de Caen Normandie, 14032 Caen, France; (A.M.); (K.B.)
| |
Collapse
|
9
|
Wu R, Wu J, Jin H, Ma H, Huang H, Xu W, Sun S, Liu X, Dong K, Xie Y, Zeng J, Wang F. Olink and gut microbial metabolomics reveal new biomarkers for the prediction and diagnosis of PMOP. J Bone Miner Metab 2024; 42:503-515. [PMID: 39153113 DOI: 10.1007/s00774-024-01545-z] [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: 03/19/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
LNTRODUCTION Postmenopausal osteoporosis (PMOP) can cause postmenopausal women to experience pain and interference. Identifying and exploring potential early diagnostic biomarkers of PMOP is of substantial clinical value and social significance. This study aimed to screen for potential novel diagnostic biomarkers of PMOP through a multiomics approach, providing new directions and ideas for the early prevention and treatment of this disease. MATERIALS AND METHODS Fifteen postmenopausal women with osteoporosis and 12 without were recruited. Clinical information was collected, and various clinical biochemical parameters were tested. Plasma and fecal samples were collected and analyzed using Olink proteomics and gut microbial metabolomics. RESULTS The functions of the differentially abundant metabolites were mainly related to autophagy and arginine and proline metabolism and were involved in immunoinflammatory metabolic processes. Olink showed significant differences in the expression of seven inflammation-related proteins between the two groups. CONCLUSION We demonstrated that metabolic differences between PMOP patients and healthy controls were associated with inflammatory responses and found seven proteins with significant differences. Among these proteins, CDCP1, IL10, and IL-1alpha combined with clinical indicators had high discriminant efficiency in identifying PMOP. This is also the first study to demonstrate noteworthy changes in CDCP1 levels in patients with PMOP.
Collapse
Affiliation(s)
- Ruizhe Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Jie Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Hui Jin
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Huaiyu Ma
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Hongxing Huang
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Wuji Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Shaoqiu Sun
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xiaolan Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Kefang Dong
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Yisong Xie
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Jingqi Zeng
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| | - Fan Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| |
Collapse
|
10
|
Traina G. Mast Cells in Human Health and Diseases 2.0. Int J Mol Sci 2024; 25:6443. [PMID: 38928149 PMCID: PMC11203736 DOI: 10.3390/ijms25126443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/04/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
This Special Issue collects some scientific pieces of the multifaceted research on the mast cell (MC), and it intends to highlight the broad spectrum of activity that MCs have, both in physiological conditions and in pathological states, focusing attention on some of them [...].
Collapse
Affiliation(s)
- Giovanna Traina
- Department of Pharmaceutical Sciences, University of Perugia, Via Romana, 06126 Perugia, Italy
| |
Collapse
|
11
|
Molitoris KH, Huang M, Baht GS. Osteoimmunology of Fracture Healing. Curr Osteoporos Rep 2024; 22:330-339. [PMID: 38616228 PMCID: PMC11186872 DOI: 10.1007/s11914-024-00869-z] [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] [Accepted: 03/26/2024] [Indexed: 04/16/2024]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize what is known in the literature about the role inflammation plays during bone fracture healing. Bone fracture healing progresses through four distinct yet overlapping phases: formation of the hematoma, development of the cartilaginous callus, development of the bony callus, and finally remodeling of the fracture callus. Throughout this process, inflammation plays a critical role in robust bone fracture healing. RECENT FINDINGS At the onset of injury, vessel and matrix disruption lead to the generation of an inflammatory response: inflammatory cells are recruited to the injury site where they differentiate, activate, and/or polarize to secrete cytokines for the purposes of cell signaling and cell recruitment. This process is altered by age and by sex. Bone fracture healing is heavily influenced by the presence of inflammatory cells and cytokines within the healing tissue.
Collapse
Affiliation(s)
- Kristin Happ Molitoris
- Department of Orthopaedic Surgery, Duke Molecular Physiology Institute, Duke University, 300 North Duke Street, Durham, NC, 27701, USA
| | - Mingjian Huang
- Department of Orthopaedic Surgery, Duke Molecular Physiology Institute, Duke University, 300 North Duke Street, Durham, NC, 27701, USA
| | - Gurpreet Singh Baht
- Department of Orthopaedic Surgery, Duke Molecular Physiology Institute, Duke University, 300 North Duke Street, Durham, NC, 27701, USA.
| |
Collapse
|
12
|
Zhou X, Chen Y, Zhang Z, Miao J, Chen G, Qian Z. Identification of differentially expressed genes, signaling pathways and immune infiltration in postmenopausal osteoporosis by integrated bioinformatics analysis. Heliyon 2024; 10:e23794. [PMID: 38205281 PMCID: PMC10777010 DOI: 10.1016/j.heliyon.2023.e23794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
Background Postmenopausal osteoporosis is a systemic metabolic disorder typified by an imbalance in bone turnover, where bone resorption supersedes bone formation. This imbalance primarily arises from a decline in bone mass induced by estrogen deficiency, and an elevated risk of fractures resulting from degradation of bone microstructure. Despite recognizing these changes, the precise causative factors and potential molecular pathways remain elusive. In this study, we aimed to identify differentially expressed genes (DEGs), associated pathways, and the role of immune infiltration in osteoporosis, leveraging an integrated bioinformatics approach to shed light on potential underlying molecular mechanisms. Methods We retrieved the expression profiles of GSE230665 from the Gene Expression Omnibus database, comprising 15 femur samples, including 12 postmenopausal osteoporosis samples and 3 normal controls. From the aggregated microarray datasets, we derived differentially expressed genes (DEGs) for further bioinformatics analysis. We used WGCNA, analyzed DEGs, PPI, and conducted GO analysis to identify pivotal genes. We then used the CIBERSORT method to explore the degree of immune cell infiltration within femur specimens affected by postmenopausal osteoporosis. To probe into the relationship between pivotal genes and infiltrating immune cells, we conducted correlation analysis. Results We identified a total of 12,204 DEGs. Among these, 12,157 were up-regulated, and 47 were down-regulated. GO and KEGG pathway analyses indicated that these DEGs predominantly targeted cellular protein localization activity and associated signaling pathways. The protein-protein interaction network highlighted four central hub-genes: RPL31, RPL34, EEF1G, and BPTF. Principal component analysis indicated a positive correlation between the expression of these genes and resting NK cells (as per CIBERSORT). In contrast, the expression of RPL31, RPL34, and EEF1G showed a negative correlation with T cells (gamma delta per CIBERSORT). Conclusions Immune infiltration plays a role in the development of osteoporosis.
Collapse
Affiliation(s)
- Xiaoli Zhou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin 300211, China
- Department of Toxicology, Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China
| | - Yang Chen
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin 300211, China
| | - Zepei Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin 300211, China
| | - Jun Miao
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin 300211, China
| | - Guangdong Chen
- Department of Orthopaedics, Cangzhou Central Hospital, Hebei 061001, China
| | - Zhiyong Qian
- Department of Toxicology, Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China
| |
Collapse
|
13
|
Zhao S, Qiao Z, Pfeifer R, Pape HC, Mao K, Tang H, Meng B, Chen S, Liu H. Modulation of fracture healing by senescence-associated secretory phenotype (SASP): a narrative review of the current literature. Eur J Med Res 2024; 29:38. [PMID: 38195489 PMCID: PMC10775505 DOI: 10.1186/s40001-023-01604-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 12/19/2023] [Indexed: 01/11/2024] Open
Abstract
The senescence-associated secretory phenotype (SASP) is a generic term for the secretion of cytokines, such as pro-inflammatory factors and proteases. It is a crucial feature of senescent cells. SASP factors induce tissue remodeling and immune cell recruitment. Previous studies have focused on the beneficial role of SASP during embryonic development, wound healing, tissue healing in general, immunoregulation properties, and cancer. However, some recent studies have identified several negative effects of SASP on fracture healing. Senolytics is a drug that selectively eliminates senescent cells. Senolytics can inhibit the function of senescent cells and SASP, which has been found to have positive effects on a variety of aging-related diseases. At the same time, recent data suggest that removing senescent cells may promote fracture healing. Here, we reviewed the latest research progress about SASP and illustrated the inflammatory response and the influence of SASP on fracture healing. This review aims to understand the role of SASP in fracture healing, aiming to provide an important clinical prevention and treatment strategy for fracture. Clinical trials of some senolytics agents are underway and are expected to clarify the effectiveness of their targeted therapy in the clinic in the future. Meanwhile, the adverse effects of this treatment method still need further study.
Collapse
Affiliation(s)
- Shangkun Zhao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhi Qiao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Roman Pfeifer
- Department of Traumatology, University Hospital of Zurich, Zurich, 8091, China
| | - Hans-Christoph Pape
- Department of Traumatology, University Hospital of Zurich, Zurich, 8091, China
| | - Keya Mao
- Chinese PLA General Hospital Beijing, Beijing, 100853, China
| | - Hai Tang
- Beijing Friendship Hospital, Beijing, 100050, China
| | - Bin Meng
- First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Songfeng Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongjian Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
14
|
Wang P, Gong Y, Zhou G, Ren W, Wang X. Biodegradable Implants for Internal Fixation of Fractures and Accelerated Bone Regeneration. ACS OMEGA 2023; 8:27920-27931. [PMID: 37576626 PMCID: PMC10413843 DOI: 10.1021/acsomega.3c02727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023]
Abstract
Bone fractures have always been a burden to patients due to their common occurrence and severe complications. Traditionally, operative treatments have been widely used in the clinic for implanting, despite the fact that they can only achieve bone fixation with limited stability and pose no effect on promoting tissue growth. In addition, the nondegradable implants usually need a secondary surgery for implant removal, otherwise they may block the regeneration of bones resulting in bone nonunion. To overcome the low degradability of implants and avoid multiple surgeries, tissue engineers have investigated various biodegradable materials for bone regeneration, whereas the significance of stability of long-term bone fixation tends to be neglected during this process. Combining the traditional orthopedic implantation surgeries and emerging tissue engineering, we believe that both bone fixation and bone regeneration are indispensable factors for a successful bone repair. Herein, we define such a novel idea as bone regenerative fixation (BRF), which should be the main future development trend of biodegradable materials.
Collapse
Affiliation(s)
- Pei Wang
- Department
of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of
Tissue Engineering, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yan Gong
- Department
of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of
Tissue Engineering, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Guangdong Zhou
- Department
of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of
Tissue Engineering, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Institute
of Regenerative Medicine and Orthopedics, Institutes of Health Central
Plain, Xinxiang Medical University, Henan 453003, China
| | - Wenjie Ren
- Institute
of Regenerative Medicine and Orthopedics, Institutes of Health Central
Plain, Xinxiang Medical University, Henan 453003, China
| | - Xiansong Wang
- Department
of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of
Tissue Engineering, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Institute
of Regenerative Medicine and Orthopedics, Institutes of Health Central
Plain, Xinxiang Medical University, Henan 453003, China
| |
Collapse
|
15
|
Fischer V, Bülow JM, Krüger BT, Ragipoglu D, Vikman A, Haffner-Luntzer M, Katsoulis-Dimitriou K, Dudeck A, Ignatius A. Role of Mast-Cell-Derived RANKL in Ovariectomy-Induced Bone Loss in Mice. Int J Mol Sci 2023; 24:ijms24119135. [PMID: 37298085 DOI: 10.3390/ijms24119135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/17/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Mast cells may contribute to osteoporosis development, because patients with age-related or post-menopausal osteoporosis exhibit more mast cells in the bone marrow, and mastocytosis patients frequently suffer from osteopenia. We previously showed that mast cells crucially regulated osteoclastogenesis and bone loss in ovariectomized, estrogen-depleted mice in a preclinical model for post-menopausal osteoporosis and found that granular mast cell mediators were responsible for these estrogen-dependent effects. However, the role of the key regulator of osteoclastogenesis, namely, receptor activator of NFκB ligand (RANKL), which is secreted by mast cells, in osteoporosis development has, to date, not been defined. Here, we investigated whether mast-cell-derived RANKL participates in ovariectomy (OVX)-induced bone loss by using female mice with a conditional Rankl deletion. We found that this deletion in mast cells did not influence physiological bone turnover and failed to protect against OVX-induced bone resorption in vivo, although we demonstrated that RANKL secretion was significantly reduced in estrogen-treated mast cell cultures. Furthermore, Rankl deletion in mast cells did not influence the immune phenotype in non-ovariectomized or ovariectomized mice. Therefore, other osteoclastogenic factors released by mast cells might be responsible for the onset of OVX-induced bone loss.
Collapse
Affiliation(s)
- Verena Fischer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, 89081 Ulm, Germany
| | - Jasmin Maria Bülow
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, 89081 Ulm, Germany
| | - Benjamin Thilo Krüger
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, 89081 Ulm, Germany
| | - Deniz Ragipoglu
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, 89081 Ulm, Germany
| | - Anna Vikman
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, 89081 Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, 89081 Ulm, Germany
| | - Konstantinos Katsoulis-Dimitriou
- Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Anne Dudeck
- Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, 89081 Ulm, Germany
| |
Collapse
|
16
|
Ghosh M, Rana S. The anaphylatoxin C5a: Structure, function, signaling, physiology, disease, and therapeutics. Int Immunopharmacol 2023; 118:110081. [PMID: 36989901 DOI: 10.1016/j.intimp.2023.110081] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
The complement system is one of the oldest known tightly regulated host defense systems evolved for efficiently functioning cell-based immune systems and antibodies. Essentially, the complement system acts as a pivot between the innate and adaptive arms of the immune system. The complement system collectively represents a cocktail of ∼50 cell-bound/soluble glycoproteins directly involved in controlling infection and inflammation. Activation of the complement cascade generates complement fragments like C3a, C4a, and C5a as anaphylatoxins. C5a is the most potent proinflammatory anaphylatoxin, which is involved in inflammatory signaling in a myriad of tissues. This review provides a comprehensive overview of human C5a in the context of its structure and signaling under several pathophysiological conditions, including the current and future therapeutic applications targeting C5a.
Collapse
Affiliation(s)
- Manaswini Ghosh
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha 752050, India
| | - Soumendra Rana
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha 752050, India.
| |
Collapse
|
17
|
Oxidative Stress and Inflammation in Osteoporosis: Molecular Mechanisms Involved and the Relationship with microRNAs. Int J Mol Sci 2023; 24:ijms24043772. [PMID: 36835184 PMCID: PMC9963528 DOI: 10.3390/ijms24043772] [Citation(s) in RCA: 124] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Osteoporosis is characterized by the alteration of bone homeostasis due to an imbalance between osteoclastic bone resorption and osteoblastic bone formation. Estrogen deficiency causes bone loss and postmenopausal osteoporosis, the pathogenesis of which also involves oxidative stress, inflammatory processes, and the dysregulation of the expression of microRNAs (miRNAs) that control gene expression at post-transcriptional levels. Oxidative stress, due to an increase in reactive oxygen species (ROS), proinflammatory mediators and altered levels of miRNAs enhance osteoclastogenesis and reduce osteoblastogenesis through mechanisms involving the activation of MAPK and transcription factors. The present review summarizes the principal molecular mechanisms involved in the role of ROS and proinflammatory cytokines on osteoporosis. Moreover, it highlights the interplay among altered miRNA levels, oxidative stress, and an inflammatory state. In fact, ROS, by activating the transcriptional factors, can affect miRNA expression, and miRNAs can regulate ROS production and inflammatory processes. Therefore, the present review should help in identifying targets for the development of new therapeutic approaches to osteoporotic treatment and improve the quality of life of patients.
Collapse
|
18
|
Shakurov AV, Lukina YS, Skriabin AS, Bionyshev-Abramov LL, Serejnikova NB, Smolencev DV. Enhanced bone healing using local cryostimulation: In vivo rat study. J Therm Biol 2023; 113:103501. [PMID: 37055120 DOI: 10.1016/j.jtherbio.2023.103501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/09/2022] [Accepted: 02/05/2023] [Indexed: 02/12/2023]
Abstract
A short-term intense cooling through the skin causes a response of the body. Potentially, it can be used to enhance bone healing. The aim of this study is to evaluate an effectiveness of a bone defect cryostimulation in the Wistar rat model in vivo. Through holes with a diameter of 2.15 mm were formed in the cortical layer of the diaphysis of the hind paws of rats. Further animals were subjected to cryotherapy 1 and 2 times a week (up to 6 weeks). The local average skin surface temperature dropped from 28 to 14 °C. The decrease in temperature in a control point inside the biological tissue was 5.3 °C. Micro CT and histological analyses showed that cryostimulation twice a week is efficient treatment. In this case, there was an acceleration of maturation of the newly formed bone tissue replacing the defect region. In the control, the newly formed immature bone with a large number of osteocytes and vessels was detected. In the experiment, the newly formed bone had a more mature structure with signs of a compact bone (formation of Haversian canals, reduction in the number of osteocytes, appearance of gluing lines). Morphometric analysis has showed a 2-fold decrease of the relative vessels area near the defect region and an increase of 30% in the content of mast cells in the entire bone marrow and especially near the site of osteogenesis. Generally, the complete filling of the critical size defect and almost complete mineralization have been observed. This information is expected to be useful for understanding the effect-exposure correlation of the cryotherapy and in the design of the cryotherapy protocols.
Collapse
Affiliation(s)
- A V Shakurov
- Bauman Moscow State Technical University (National Research University), Moscow, 105005, Baumanskaya 2-ya St., 5, Russian Federation.
| | - Yu S Lukina
- Bauman Moscow State Technical University (National Research University), Moscow, 105005, Baumanskaya 2-ya St., 5, Russian Federation; National Medical Research Center for Traumatology and Orthopedics Named After N.N. Priorov, Ministry of Health of the Russian Federation, Moscow, 127299, Priorova St., 10, Russian Federation
| | - A S Skriabin
- Bauman Moscow State Technical University (National Research University), Moscow, 105005, Baumanskaya 2-ya St., 5, Russian Federation
| | - L L Bionyshev-Abramov
- National Medical Research Center for Traumatology and Orthopedics Named After N.N. Priorov, Ministry of Health of the Russian Federation, Moscow, 127299, Priorova St., 10, Russian Federation
| | - N B Serejnikova
- National Medical Research Center for Traumatology and Orthopedics Named After N.N. Priorov, Ministry of Health of the Russian Federation, Moscow, 127299, Priorova St., 10, Russian Federation; Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Trubetskaya St., 8, Russian Federation
| | - D V Smolencev
- National Medical Research Center for Traumatology and Orthopedics Named After N.N. Priorov, Ministry of Health of the Russian Federation, Moscow, 127299, Priorova St., 10, Russian Federation
| |
Collapse
|
19
|
Duda GN, Geissler S, Checa S, Tsitsilonis S, Petersen A, Schmidt-Bleek K. The decisive early phase of bone regeneration. Nat Rev Rheumatol 2023; 19:78-95. [PMID: 36624263 DOI: 10.1038/s41584-022-00887-0] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2022] [Indexed: 01/11/2023]
Abstract
Bone has a remarkable endogenous regenerative capacity that enables scarless healing and restoration of its prior mechanical function, even under challenging conditions such as advanced age and metabolic or immunological degenerative diseases. However - despite much progress - a high number of bone injuries still heal with unsatisfactory outcomes. The mechanisms leading to impaired healing are heterogeneous, and involve exuberant and non-resolving immune reactions or overstrained mechanical conditions that affect the delicate regulation of the early initiation of scar-free healing. Every healing process begins phylogenetically with an inflammatory reaction, but its spatial and temporal intensity must be tightly controlled. Dysregulation of this inflammatory cascade directly affects the subsequent healing phases and hinders the healing progression. This Review discusses the complex processes underlying bone regeneration, focusing on the early healing phase and its highly dynamic environment, where vibrant changes in cellular and tissue composition alter the mechanical environment and thus affect the signalling pathways that orchestrate the healing process. Essential to scar-free healing is the interplay of various dynamic cascades that control timely resolution of local inflammation and tissue self-organization, while also providing sufficient local stability to initiate endogenous restoration. Various immunotherapy and mechanobiology-based therapy options are under investigation for promoting bone regeneration.
Collapse
Affiliation(s)
- Georg N Duda
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany. .,Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Sven Geissler
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Serafeim Tsitsilonis
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ansgar Petersen
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
20
|
Chang B, Hu Z, Chen L, Jin Z, Yang Y. Development and validation of cuproptosis-related genes in synovitis during osteoarthritis progress. Front Immunol 2023; 14:1090596. [PMID: 36817415 PMCID: PMC9932029 DOI: 10.3389/fimmu.2023.1090596] [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: 11/09/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common refractory degenerative joint diseases worldwide. Synovitis is believed to drive joint cartilage destruction during OA pathogenesis. Cuproptosis is a novel form of copper-induced cell death. However, few studies have examined the correlations between cuproptosis-related genes (CRGs), immune infiltration, and synovitis. Therefore, we analyzed CRGs in synovitis during OA. Microarray datasets (GSE55235, GSE55457, GSE12021, GSE82107 and GSE176308) were downloaded from the Gene Expression Omnibus database. Next, we conducted differential and subtype analyses of CRGs across synovitis. Immune infiltration and correlation analyses were performed to explore the association between CRGs and immune cell abundance in synovitis. Finally, single-cell RNA-seq profiling was performed using the GSE176308 dataset to investigate the expression of CRGs in the various cell clusters. We found that the expression of five CRGs (FDX1, LIPT1, PDHA1, PDHB, and CDKN2A) was significantly increased in the OA synovium. Moreover, abundant and various types of immune cells infiltrated the synovium during OA, which was correlated with the expression of CRGs. Additionally, single-cell RNA-seq profiling revealed that the cellular composition of the synovium was complex and that their proportions varied greatly as OA progressed. The expression of CRGs differed across various cell types in the OA synovium. The current study predicted that cuproptosis may be involved in the pathogenesis of synovitis. The five screened CRGs (FDX1, LIPT1, PDHA1, PDHB, and CDKN2A) could be explored as candidate biomarkers or therapeutic targets for OA synovitis.
Collapse
Affiliation(s)
- Bohan Chang
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhehan Hu
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liang Chen
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhuangzhuang Jin
- Department of Emergence Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Yang
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
21
|
Ma C, Li H, Lu S, Li X, Wang S, Wang W. Tryptase and Exogenous Trypsin: Mechanisms and Ophthalmic Applications. J Inflamm Res 2023; 16:927-939. [PMID: 36891173 PMCID: PMC9987324 DOI: 10.2147/jir.s402900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Ocular injuries caused by inflammation, surgery or accidents are subject to a physiological healing process that ultimately restores the structure and function of the damaged tissue. Tryptase and trypsin are essential component of this process and they play a role in promoting and reducing the inflammatory response of tissues, respectively. Following injury, tryptase is endogenously produced by mast cells and can exacerbate the inflammatory response both by stimulating neutrophil secretion, and through its agonist action on proteinase-activated receptor 2 (PAR2). In contrast, exogenously introduced trypsin promotes wound healing by attenuating inflammatory responses, reducing oedema and protecting against infection. Thus, trypsin may help resolve ocular inflammatory symptoms and promote faster recovery from acute tissue injury associated with ophthalmic diseases. This article describes the roles of tryptase and exogenous trypsin in affected tissues after onset of ocular injury, and the clinical applications of trypsin injection.
Collapse
Affiliation(s)
- Chao Ma
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Haoyu Li
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China.,Hunan Clinical Research Centre of Ophthalmic Disease, Changsha, Hunan, People's Republic of China
| | - Shuwen Lu
- Department of Ophthalmology, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, People's Republic of China
| | - Xian Li
- Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester, UK.,Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, the University of Manchester, Manchester, UK
| | - Shuai Wang
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Wenzhan Wang
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
22
|
Haffner-Luntzer M, Weber B, Morioka K, Lackner I, Fischer V, Bahney C, Ignatius A, Kalbitz M, Marcucio R, Miclau T. Altered early immune response after fracture and traumatic brain injury. Front Immunol 2023; 14:1074207. [PMID: 36761764 PMCID: PMC9905106 DOI: 10.3389/fimmu.2023.1074207] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
Introduction Clinical and preclinical data suggest accelerated bone fracture healing in subjects with an additional traumatic brain injury (TBI). Mechanistically, altered metabolism and neuro-endocrine regulations have been shown to influence bone formation after combined fracture and TBI, thereby increasing the bone content in the fracture callus. However, the early inflammatory response towards fracture and TBI has not been investigated in detail so far. This is of great importance, since the early inflammatory phase of fracture healing is known to be essential for the initiation of downstream regenerative processes for adequate fracture repair. Methods Therefore, we analyzed systemic and local inflammatory mediators and immune cells in mice which were exposed to fracture only or fracture + TBI 6h and 24h after injury. Results We found a dysregulated systemic immune response and significantly fewer neutrophils and mast cells locally in the fracture hematoma. Further, local CXCL10 expression was significantly decreased in the animals with combined trauma, which correlated significantly with the reduced mast cell numbers. Discussion Since mast cells and mast cell-derived CXCL10 have been shown to increase osteoclastogenesis, the reduced mast cell numbers might contribute to higher bone content in the fracture callus of fracture + TBI mice due to decreased callus remodeling.
Collapse
Affiliation(s)
- Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany.,Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Birte Weber
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany.,Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Kazuhito Morioka
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Ina Lackner
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Chelsea Bahney
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,Steadman Phillipon Research Institute, Vail, CO, United States
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
23
|
Local immune cell contributions to fracture healing in aged individuals - A novel role for interleukin 22. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1262-1276. [PMID: 36028760 PMCID: PMC9440089 DOI: 10.1038/s12276-022-00834-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/25/2022] [Accepted: 06/06/2022] [Indexed: 11/08/2022]
Abstract
With increasing age, the risk of bone fractures increases while regenerative capacity decreases. This variation in healing potential appears to be linked to adaptive immunity, but the underlying mechanism is still unknown. This study sheds light on immunoaging/inflammaging, which impacts regenerative processes in aging individuals. In an aged preclinical model system, different levels of immunoaging were analyzed to identify key factors that connect immunoaged/inflammaged conditions with bone formation after long bone fracture. Immunological facets, progenitor cells, the microbiome, and confounders were monitored locally at the injury site and systemically in relation to healing outcomes in 12-month-old mice with distinct individual levels of immunoaging. Bone tissue formation during healing was delayed in the immunoaged group and could be associated with significant changes in cytokine levels. A prolonged and amplified pro-inflammatory reaction was caused by upregulated immune cell activation markers, increased chemokine receptor availability and a lack of inhibitory signaling. In immunoaged mice, interleukin-22 was identified as a core cell signaling protein that played a central role in delayed healing. Therapeutic neutralization of IL-22 reversed this specific immunoaging-related disturbed healing. Immunoaging was found to be an influencing factor of decreased regenerative capacity in aged individuals. Furthermore, a novel therapeutic strategy of neutralizing IL-22 may successfully rejuvenate healing in individuals with advanced immune experiences.
Collapse
|
24
|
Ruocco A, Sirico A, Novelli R, Iannelli S, Van Breda SV, Kyburz D, Hasler P, Aramini A, Amendola PG. The role of C5a-C5aR1 axis in bone pathophysiology: A mini-review. Front Cell Dev Biol 2022; 10:957800. [PMID: 36003145 PMCID: PMC9393612 DOI: 10.3389/fcell.2022.957800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Bone remodeling is a physiological, dynamic process that mainly depends on the functions of 2 cell types: osteoblasts and osteoclasts. Emerging evidence suggests that complement system is crucially involved in the regulation of functions of these cells, especially during inflammatory states. In this context, complement component 5a (C5a), a powerful pro-inflammatory anaphylatoxin that binds the receptor C5aR1, is known to regulate osteoclast formation and osteoblast inflammatory responses, and has thus been proposed as potential therapeutic target for the treatment of inflammatory bone diseases. In this review, we will analyze the role of C5a-C5aR1 axis in bone physiology and pathophysiology, describing its involvement in the pathogenesis of some of the most frequent inflammatory bone diseases such as rheumatoid arthritis, and also in osteoporosis and bone cancer and metastasis. Moreover, we will examine C5aR1-based pharmacological approaches that are available and have been tested so far for the treatment of these conditions. Given the growing interest of the scientific community on osteoimmunology, and the scarcity of data regarding the role of C5a-C5aR1 axis in bone pathophysiology, we will highlight the importance of this axis in mediating the interactions between skeletal and immune systems and its potential use as a therapeutic target.
Collapse
Affiliation(s)
| | | | | | | | | | - Diego Kyburz
- Departement Biomedizin, University of Basel, Basel, Switzerland
| | - Paul Hasler
- Division of Rheumatology, Kantonsspital Aarau AG, Aarau, Switzerland
| | | | | |
Collapse
|
25
|
Ng CW, Chan BCL, Ko CH, Tam IYS, Sam SW, Lau CBS, Leung PC, Lau HYA. Human mast cells induce osteoclastogenesis through cell surface RANKL. Inflamm Res 2022; 71:1261-1270. [PMID: 35916930 DOI: 10.1007/s00011-022-01608-9] [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] [Accepted: 07/05/2022] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES We employed the co-culture of CD34+ stem cell-derived human mast cells (HMC) and human monocyte-derived osteoclast precursors to evaluate if mast cells contribute to the pathogenesis of osteoporosis through regulation of osteoclast proliferation and activation. METHODS Mature HMC and osteoclast precursors were cultured from monocytes isolated from human buffy coat. The osteoclast precursors were incubated with HMC or receptor activator of nuclear factor kappa-B ligand (RANKL) for a week prior to determination of osteoclast maturation through characterization by their morphology and tartrate resistant acid phosphatase (TRAP) expression. The bone absorption activity was determined by pit formation on osteo-assay plate. RESULTS Mature osteoclasts were identified following co-culture of osteoclast precursors with HMC for one week in the absence of RANKL and they were capable of bone resorption. These actions of HMC on osteoclasts were not affected by mast cell activators such anti-IgE or substance P but could be reversed by osteoprotegerin (OPG) in the co-culture system suggesting the involvement of RANKL. The expression of RANKL on the cell surface of HMC was confirmed by flow cytometry and the density was not affected by activation of HMC. CONCLUSION Our study provided direct evidence confirming the initiation of osteoclast proliferation and activation by mast cells through cell surface RANKL suggesting that mast cells may contribute to bone destruction in pathological conditions such as osteoporosis.
Collapse
Affiliation(s)
- Chun Wai Ng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Ben Chung Lap Chan
- Institute of Chinese Medicine and State Key Laboratory of Research On Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Chun Hay Ko
- Institute of Chinese Medicine and State Key Laboratory of Research On Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Issan Yee San Tam
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Sze Wing Sam
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Clara Bik San Lau
- Institute of Chinese Medicine and State Key Laboratory of Research On Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Ping Chung Leung
- Institute of Chinese Medicine and State Key Laboratory of Research On Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Hang Yung Alaster Lau
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
26
|
Livshits G, Kalinkovich A. Targeting chronic inflammation as a potential adjuvant therapy for osteoporosis. Life Sci 2022; 306:120847. [PMID: 35908619 DOI: 10.1016/j.lfs.2022.120847] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/07/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022]
Abstract
Systemic, chronic, low-grade inflammation (SCLGI) underlies the pathogenesis of various widespread diseases. It is often associated with bone loss, thus connecting chronic inflammation to the pathogenesis of osteoporosis. In postmenopausal women, osteoporosis is accompanied by SCLGI development, likely owing to estrogen deficiency. We propose that SCGLI persistence in osteoporosis results from failed inflammation resolution, which is mainly mediated by specialized, pro-resolving mediators (SPMs). In corroboration, SPMs demonstrate encouraging therapeutic effects in various preclinical models of inflammatory disorders, including bone pathology. Since numerous data implicate gut dysbiosis in osteoporosis-associated chronic inflammation, restoring balanced microbiota by supplementing probiotics and prebiotics could contribute to the efficient resolution of SCGLI. In the present review, we provide evidence for this hypothesis and argue that efficient SCGLI resolution may serve as a novel approach for treating osteoporosis, complementary to traditional anti-osteoporotic medications.
Collapse
Affiliation(s)
- Gregory Livshits
- Adelson School of Medicine, Ariel University, Ariel 4077625, Israel; Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel.
| | - Alexander Kalinkovich
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel
| |
Collapse
|
27
|
Song J, Hu L, Liu B, Jiang N, Huang H, Luo J, Wang L, Zeng J, Huang F, Huang M, Cai L, Tang L, Chen S, Chen Y, Wu A, Zheng S, Chen Q. The Emerging Role of Immune Cells and Targeted Therapeutic Strategies in Diabetic Wounds Healing. J Inflamm Res 2022; 15:4119-4138. [PMID: 35898820 PMCID: PMC9309318 DOI: 10.2147/jir.s371939] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/07/2022] [Indexed: 11/23/2022] Open
Abstract
Poor wound healing in individuals with diabetes has long plagued clinicians, and immune cells play key roles in the inflammation, proliferation and remodeling that occur in wound healing. When skin integrity is damaged, immune cells migrate to the wound bed through the actions of chemokines and jointly restore tissue homeostasis and barrier function by exerting their respective biological functions. An imbalance of immune cells often leads to ineffective and disordered inflammatory responses. Due to the maladjusted microenvironment, the wound is unable to smoothly transition to the proliferation and remodeling stage, causing it to develop into a chronic refractory wound. However, chronic refractory wounds consistently lead to negative outcomes, such as long treatment cycles, high hospitalization rates, high medical costs, high disability rates, high mortality rates, and many adverse consequences. Therefore, strategies that promote the rational distribution and coordinated development of immune cells during wound healing are very important for the treatment of diabetic wounds (DW). Here, we explored the following aspects by performing a literature review: 1) the current situation of DW and an introduction to the biological functions of immune cells; 2) the role of immune cells in DW; and 3) existing (or undeveloped) therapies targeting immune cells to promote wound healing to provide new ideas for basic research, clinical treatment and nursing of DW.
Collapse
Affiliation(s)
- Jianying Song
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Lixin Hu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Bo Liu
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Nan Jiang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Houqiang Huang
- Department of Nursing, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - JieSi Luo
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Long Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Jing Zeng
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Feihong Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Min Huang
- Department of Respiratory and Critical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Luyao Cai
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Lingyu Tang
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Shunli Chen
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Yinyi Chen
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Anguo Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Silin Zheng
- Department of Nursing, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Qi Chen
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| |
Collapse
|
28
|
Does anti-IgE therapy prevent chronic allergic asthma-related bone deterioration in asthmatic mice? J Biomech 2022; 141:111180. [PMID: 35724549 DOI: 10.1016/j.jbiomech.2022.111180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 11/21/2022]
Abstract
Current evidence on the association between allergic diseases and bone metabolism indicates asthma may be a potential risk factor for bone health. Using anti-IgE has been proven effective in allergic asthma treatment with a good safety profile; however, its effects on bone health are unknown. Thus, we aimed to investigate whether: (i) chronic allergic asthma (CAA) causes any meaningful changes in bone, and if any, (ii) anti-IgE therapy prevents any CAA-induced adverse alteration. A murine model was used to study CAA. Thirty-two BALB/c male-mice were assigned into four groups (eight-mice/group): Control, CAA (treated with saline), CAA + 100 µg of anti-IgE (CAA + 100AIgE), and CAA + 200 µg of anti-IgE (CAA + 200AIgE) groups. After immunization, saline or anti-IgE was performed intraperitoneally for 8-weeks (in five-sessions at 15-days interval). Three-point bending test was used for the mechanical analysis. Bone calcium (Ca2+) and phosphorus (P3-) as well as Ca/P ratio were evaluated using inductively-coupled plasma-mass-spectrometer (ICP-MS). Compared to control, reductions observed in yield and ultimate moments, rigidity, energy-to-failure, yield and ultimate stresses, elastic modulus, toughness, and post-yield toughness parameters of the CAA group were found significant (P < 0.05). Similar declines were also detected regarding bone Ca2+, P3- and Ca/P ratio (P < 0.05). Compared to control, we observed that 200 µg administration of anti-IgE in CAA + 200AIgE group hindered CAA-related impairments in mineral and mechanical characteristics of bone, while 100 µg in CAA + 100AIgE failed to do so. Our results showed CAA may cause bone loss, leading to a decrease in bone strength, and anti-IgE administration may dose-dependently inhibit these impairments in bone.
Collapse
|
29
|
Ragipoglu D, Bülow J, Hauff K, Voss M, Haffner-Luntzer M, Dudeck A, Ignatius A, Fischer V. Mast Cells Drive Systemic Inflammation and Compromised Bone Repair After Trauma. Front Immunol 2022; 13:883707. [PMID: 35558068 PMCID: PMC9086903 DOI: 10.3389/fimmu.2022.883707] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
There is evidence that mast cells contribute to inflammation induced by hemorrhagic shock, severe tissue injury or sepsis. Mast cells are highly responsive to alarm signals generated after trauma, and release many inflammatory mediators including interleukin-6, a key mediator of posttraumatic inflammation. An overwhelming posttraumatic inflammation causes compromised bone healing; however, the underlying cellular and molecular mechanisms are poorly understood. Recently, we found that mast cells trigger local and systemic inflammation after isolated fracture leading to uneventful bone repair. Here, we investigated whether mast cells critically contribute to trauma-induced compromised bone healing. Male Mcpt5-Cre+ R-DTA mice, which lack connective tissue type mast cells, and their mast cell-competent Cre- littermates underwent a femur fracture with/without thoracic trauma. Posttraumatic systemic and local inflammation and bone repair were assessed 3 h and 21 d post injury. Both, the systemic and pulmonary inflammation was significantly increased in mast cell-competent mice upon combined trauma compared to isolated fracture. In mast cell-deficient mice, the increase of inflammatory mediators in the circulation induced by the severe trauma was abolished. In the bronchoalveolar lavage fluid, the trauma-induced increase of inflammatory cytokines was not reduced, but the neutrophil invasion into the lungs was significantly diminished in the absence of mast cells. Locally in the fracture hematoma, mast cell-competent mice displayed reduced inflammatory mediator concentrations after combined trauma compared to isolated fracture, which was abolished in mast cell-deficient mice. Notably, while combined trauma resulted in compromised bone repair in mast cell-competent mice, indicated by significantly reduced bone and increased cartilage fracture callus contents, this was abolished in Mcpt5-Cre+ R-DTA mice. Therefore, mast cells contribute to trauma-induced compromised bone repair and could be a potential target for new treatment options to improve fracture healing in multiply injured patients.
Collapse
Affiliation(s)
- Deniz Ragipoglu
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Jasmin Bülow
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Kristin Hauff
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Martin Voss
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Anne Dudeck
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
30
|
Ramirez-GarciaLuna JL, Rangel-Berridi K, Olasubulumi OO, Rosenzweig DH, Henderson JE, Gawri R, Martineau PA. Enhanced Bone Remodeling After Fracture Priming. Calcif Tissue Int 2022; 110:349-366. [PMID: 34668029 DOI: 10.1007/s00223-021-00921-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 10/01/2021] [Indexed: 11/30/2022]
Abstract
The immune system is an active component of bone repair. Mast cells influence the recruitment of macrophages, osteoclasts and blood vessels into the repair tissue. We hypothesized that if mast cells and other immune cells are sensitized to recognize broken bone, they will mount an increased response to subsequent fractures that may be translated into enhanced healing. To test this, we created a bone defect on the left leg of anesthetized mice and 2 weeks later, a second one on the right leg. Bone repair in the right legs was then compared to control mice that underwent the creation of bilateral window bone defects at the same time. Mice were euthanized at 14 and 56 days. Mineralized tissue quantity and morphometric parameters were assessed using micro-CT and histology. The activity of osteoblasts, osteoclasts, vascular endothelial cells, mast cells, and macrophages was evaluated using histochemistry. Our main findings were (1) no significant differences in the amount of bone produced at 14- or 56 days post-operative between groups; (2) mice exposed to subsequent fractures showed significantly better bone morphometric parameters after 56 days post-operative; and (3) significant increases in the content of blood vessels, osteoclasts, and the number of macrophages in the subsequent fracture group. Our results provide strong evidence that a transient increase in the inflammatory state of a healing injury promotes faster bone remodelling and increased neo-angiogenesis. This phenomenon is also characterized by changes in mast cell and macrophage content that translate into more active recruitment of mesenchymal stromal cells.
Collapse
Affiliation(s)
- Jose L Ramirez-GarciaLuna
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Karla Rangel-Berridi
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Biofabrication and Bioengineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Ore-Oluwa Olasubulumi
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
| | - Derek H Rosenzweig
- Biofabrication and Bioengineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Janet E Henderson
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
- Experimental Medicine, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| | - Rahul Gawri
- Regenerative Orthopaedics and Innovation Laboratory, Injury, Repair & Recovery Program, Research Institute-McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada.
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada.
| | - Paul A Martineau
- Bone Engineering Labs, Injury, Repair & Recovery Program, Research Institute, McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Regenerative Orthopaedics and Innovation Laboratory, Injury, Repair & Recovery Program, Research Institute-McGill University Health Centre, 1650 Cedar Ave., Montreal, QC, H3G 1A4, Canada
- Experimental Surgery, Faculty of Medicine, McGill University, 3605 Rue de la Montagne, Montreal, QC, H3G 2M1, Canada
| |
Collapse
|
31
|
Srivastava RK, Sapra L. The Rising Era of “Immunoporosis”: Role of Immune System in the Pathophysiology of Osteoporosis. J Inflamm Res 2022; 15:1667-1698. [PMID: 35282271 PMCID: PMC8906861 DOI: 10.2147/jir.s351918] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/10/2022] [Indexed: 12/21/2022] Open
Abstract
Discoveries in the last few years have emphasized the existence of an enormous breadth of communication between bone and the immune system in maintaining skeletal homeostasis. Originally, the discovery of various factors was assigned to the immune system viz. interleukin (IL)-6, IL-10, IL-17, tumor necrosis factor (TNF)-α, receptor activator of nuclear factor kappa B ligand (RANKL), nuclear factor of activated T cells (NFATc1), etc., but now these factors have also been shown to have a significant impact on osteoblasts (OBs) and osteoclasts (OCs) biology. These discoveries led to an alteration in the approach for the treatment of several bone pathologies including osteoporosis. Osteoporosis is an inflammatory bone anomaly affecting more than 500 million people globally. In 2018, to highlight the importance of the immune system in the pathophysiology of osteoporosis, our group coined the term “immunoporosis”. In the present review, we exhaustively revisit the characteristics, mechanism of action, and function of both innate and adaptive immune cells with the goal of understanding the potential of immune cells in osteoporosis. We also highlight the Immunoporotic role of gut microbiota (GM) for the treatment and management of osteoporosis. Importantly, we further discuss whether an immune cell-based strategy to treat and manage osteoporosis is feasible and relevant in clinical settings.
Collapse
Affiliation(s)
- Rupesh K Srivastava
- Immunoporosis Lab, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
- Correspondence: Rupesh K Srivastava, Tel +91 11-26593548, Email ;
| | - Leena Sapra
- Immunoporosis Lab, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| |
Collapse
|
32
|
Dahlin JS, Maurer M, Metcalfe DD, Pejler G, Sagi‐Eisenberg R, Nilsson G. The ingenious mast cell: Contemporary insights into mast cell behavior and function. Allergy 2022; 77:83-99. [PMID: 33955017 DOI: 10.1111/all.14881] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022]
Abstract
Mast cells are (in)famous for their role in allergic diseases, but the physiological and pathophysiological roles of this ingenious cell are still not fully understood. Mast cells are important for homeostasis and surveillance of the human system, recognizing both endogenous and exogenous agents, which induce release of a variety of mediators acting on both immune and non-immune cells, including nerve cells, fibroblasts, endothelial cells, smooth muscle cells, and epithelial cells. During recent years, clinical and experimental studies on human mast cells, as well as experiments using animal models, have resulted in many discoveries that help decipher the function of mast cells in health and disease. In this review, we focus particularly on new insights into mast cell biology, with a focus on mast cell development, recruitment, heterogeneity, and reactivity. We also highlight the development in our understanding of mast cell-driven diseases and discuss the development of novel strategies to treat such conditions.
Collapse
Affiliation(s)
- Joakim S. Dahlin
- Division of Immunology and Allergy Department of Medicine Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | - Marcus Maurer
- Department of Dermatology and Allergy Dermatological Allergology Allergie‐Centrum‐Charité Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, Berlin Institute of Health Berlin Germany
| | - Dean D. Metcalfe
- Mast Cell Biology Section Laboratory of Allergic Diseases NIAID, NIH Bethesda MD USA
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology Uppsala University Uppsala Sweden
- Department of Anatomy, Physiology and Biochemistry Swedish University of Agricultural Sciences Uppsala Sweden
| | - Ronit Sagi‐Eisenberg
- Department of Cell and Developmental Biology Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
| | - Gunnar Nilsson
- Division of Immunology and Allergy Department of Medicine Karolinska Institutet Karolinska University Hospital Stockholm Sweden
- Department of Medical Sciences Uppsala University Uppsala Sweden
| |
Collapse
|
33
|
Fischer V, Ragipoglu D, Diedrich J, Steppe L, Dudeck A, Schütze K, Kalbitz M, Gebhard F, Haffner-Luntzer M, Ignatius A. Mast Cells Trigger Disturbed Bone Healing in Osteoporotic Mice. J Bone Miner Res 2022; 37:137-151. [PMID: 34633111 DOI: 10.1002/jbmr.4455] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/21/2021] [Accepted: 10/03/2021] [Indexed: 12/16/2022]
Abstract
Mast cells are important tissue-resident sensor and effector immune cells but also play a major role in osteoporosis development. Mast cells are increased in numbers in the bone marrow of postmenopausal osteoporotic patients, and mast cell-deficient mice are protected from ovariectomy (OVX)-induced bone loss. In this study, we showed that mast cell-deficient Mcpt5-Cre R-DTA mice were protected from OVX-induced disturbed fracture healing, indicating a critical role for mast cells in the pathomechanisms of impaired bone repair under estrogen-deficient conditions. We revealed that mast cells trigger the fracture-induced inflammatory response by releasing inflammatory mediators, including interleukin-6, midkine (Mdk), and C-X-C motif chemokine ligand 10 (CXCL10), and promote neutrophil infiltration into the fracture site in OVX mice. Furthermore, mast cells were responsible for reduced osteoblast and increased osteoclast activities in OVX mice callus, as well as increased receptor activator of NF-κB ligand serum levels in OVX mice. Additional in vitro studies with human cells showed that mast cells stimulate osteoclastogenesis by releasing the osteoclastogenic mediators Mdk and CXCL10 in an estrogen-dependent manner, which was mediated via the estrogen receptor alpha on mast cells. In conclusion, mast cells negatively affect the healing of bone fractures under estrogen-deficient conditions. Hence, targeting mast cells might provide a therapeutic strategy to improve disturbed bone repair in postmenopausal osteoporosis. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Verena Fischer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Deniz Ragipoglu
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Johanna Diedrich
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Lena Steppe
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Anne Dudeck
- Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Konrad Schütze
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany.,Department of Trauma and Orthopedic Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen-Nürnberg, Germany
| | - Florian Gebhard
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
34
|
Ehnert S, Relja B, Schmidt-Bleek K, Fischer V, Ignatius A, Linnemann C, Rinderknecht H, Huber-Lang M, Kalbitz M, Histing T, Nussler AK. Effects of immune cells on mesenchymal stem cells during fracture healing. World J Stem Cells 2021; 13:1667-1695. [PMID: 34909117 PMCID: PMC8641016 DOI: 10.4252/wjsc.v13.i11.1667] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/31/2021] [Accepted: 09/03/2021] [Indexed: 02/06/2023] Open
Abstract
In vertebrates, bone is considered an osteoimmune system which encompasses functions of a locomotive organ, a mineral reservoir, a hormonal organ, a stem cell pool and a cradle for immune cells. This osteoimmune system is based on cooperatively acting bone and immune cells, cohabitating within the bone marrow. They are highly interdependent, a fact that is confounded by shared progenitors, mediators, and signaling pathways. Successful fracture healing requires the participation of all the precursors, immune and bone cells found in the osteoimmune system. Recent evidence demonstrated that changes of the immune cell composition and function may negatively influence bone healing. In this review, first the interplay between different immune cell types and osteoprogenitor cells will be elaborated more closely. The separate paragraphs focus on the specific cell types, starting with the cells of the innate immune response followed by cells of the adaptive immune response, and the complement system as mediator between them. Finally, a brief overview on the challenges of preclinical testing of immune-based therapeutic strategies to support fracture healing will be given.
Collapse
Affiliation(s)
- Sabrina Ehnert
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute and Berlin Institute of Health Center of Regenerative Therapies, Charité - University Medicine Berlin, Berlin 13353, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm 89091, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm 89091, Germany
| | - Caren Linnemann
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Helen Rinderknecht
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology (ITI), University Hospital Ulm, Ulm 89091, Germany
| | - Miriam Kalbitz
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen 91054, Germany
| | - Tina Histing
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Andreas K Nussler
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| |
Collapse
|
35
|
Saxena Y, Routh S, Mukhopadhaya A. Immunoporosis: Role of Innate Immune Cells in Osteoporosis. Front Immunol 2021; 12:687037. [PMID: 34421899 PMCID: PMC8374941 DOI: 10.3389/fimmu.2021.687037] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis or porous bone disorder is the result of an imbalance in an otherwise highly balanced physiological process known as 'bone remodeling'. The immune system is intricately involved in bone physiology as well as pathologies. Inflammatory diseases are often correlated with osteoporosis. Inflammatory mediators such as reactive oxygen species (ROS), and pro-inflammatory cytokines and chemokines directly or indirectly act on the bone cells and play a role in the pathogenesis of osteoporosis. Recently, Srivastava et al. (Srivastava RK, Dar HY, Mishra PK. Immunoporosis: Immunology of Osteoporosis-Role of T Cells. Frontiers in immunology. 2018;9:657) have coined the term "immunoporosis" to emphasize the role of immune cells in the pathology of osteoporosis. Accumulated pieces of evidence suggest both innate and adaptive immune cells contribute to osteoporosis. However, innate cells are the major effectors of inflammation. They sense various triggers to inflammation such as pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), cellular stress, etc., thus producing pro-inflammatory mediators that play a critical role in the pathogenesis of osteoporosis. In this review, we have discussed the role of the innate immune cells in great detail and divided these cells into different sections in a systemic manner. In the beginning, we talked about cells of the myeloid lineage, including macrophages, monocytes, and dendritic cells. This group of cells explicitly influences the skeletal system by the action of production of pro-inflammatory cytokines and can transdifferentiate into osteoclast. Other cells of the myeloid lineage, such as neutrophils, eosinophils, and mast cells, largely impact osteoporosis via the production of pro-inflammatory cytokines. Further, we talked about the cells of the lymphoid lineage, including natural killer cells and innate lymphoid cells, which share innate-like properties and play a role in osteoporosis. In addition to various innate immune cells, we also discussed the impact of classical pro-inflammatory cytokines on osteoporosis. We also highlighted the studies regarding the impact of physiological and metabolic changes in the body, which results in chronic inflammatory conditions such as ageing, ultimately triggering osteoporosis.
Collapse
Affiliation(s)
- Yogesh Saxena
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Sanjeev Routh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Arunika Mukhopadhaya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| |
Collapse
|
36
|
Li Z, Huang Z, Bai L. Cell Interplay in Osteoarthritis. Front Cell Dev Biol 2021; 9:720477. [PMID: 34414194 PMCID: PMC8369508 DOI: 10.3389/fcell.2021.720477] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/14/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a common chronic disease and a significant health concern that needs to be urgently solved. OA affects the cartilage and entire joint tissues, including the subchondral bone, synovium, and infrapatellar fat pads. The physiological and pathological changes in these tissues affect the occurrence and development of OA. Understanding complex crosstalk among different joint tissues and their roles in OA initiation and progression is critical in elucidating the pathogenic mechanism of OA. In this review, we begin with an overview of the role of chondrocytes, synovial cells (synovial fibroblasts and macrophages), mast cells, osteoblasts, osteoclasts, various stem cells, and engineered cells (induced pluripotent stem cells) in OA pathogenesis. Then, we discuss the various mechanisms by which these cells communicate, including paracrine signaling, local microenvironment, co-culture, extracellular vesicles (exosomes), and cell tissue engineering. We particularly focus on the therapeutic potential and clinical applications of stem cell-derived extracellular vesicles, which serve as modulators of cell-to-cell communication, in the field of regenerative medicine, such as cartilage repair. Finally, the challenges and limitations related to exosome-based treatment for OA are discussed. This article provides a comprehensive summary of key cells that might be targets of future therapies for OA.
Collapse
Affiliation(s)
- Zihao Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ziyu Huang
- Foreign Languages College, Shanghai Normal University, Shanghai, China
| | - Lunhao Bai
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
37
|
Jiménez M, Cervantes-García D, Córdova-Dávalos LE, Pérez-Rodríguez MJ, Gonzalez-Espinosa C, Salinas E. Responses of Mast Cells to Pathogens: Beneficial and Detrimental Roles. Front Immunol 2021; 12:685865. [PMID: 34211473 PMCID: PMC8240065 DOI: 10.3389/fimmu.2021.685865] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/28/2021] [Indexed: 12/19/2022] Open
Abstract
Mast cells (MCs) are strategically located in tissues close to the external environment, being one of the first immune cells to interact with invading pathogens. They are long living effector cells equipped with different receptors that allow microbial recognition. Once activated, MCs release numerous biologically active mediators in the site of pathogen contact, which induce vascular endothelium modification, inflammation development and extracellular matrix remodeling. Efficient and direct antimicrobial mechanisms of MCs involve phagocytosis with oxidative and non-oxidative microbial destruction, extracellular trap formation, and the release of antimicrobial substances. MCs also contribute to host defense through the attraction and activation of phagocytic and inflammatory cells, shaping the innate and adaptive immune responses. However, as part of their response to pathogens and under an impaired, sustained, or systemic activation, MCs may contribute to tissue damage. This review will focus on the current knowledge about direct and indirect contribution of MCs to pathogen clearance. Antimicrobial mechanisms of MCs are addressed with special attention to signaling pathways involved and molecular weapons implicated. The role of MCs in a dysregulated host response that can increase morbidity and mortality is also reviewed and discussed, highlighting the complexity of MCs biology in the context of host-pathogen interactions.
Collapse
Affiliation(s)
- Mariela Jiménez
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Daniel Cervantes-García
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico.,Cátedras CONACYT, National Council of Science and Technology, Mexico City, Mexico
| | - Laura E Córdova-Dávalos
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Marian Jesabel Pérez-Rodríguez
- Department of Pharmacobiology, Centro de Investigación y de Estudios Avanzados (Cinvestav), Unidad Sede Sur, Mexico City, Mexico
| | - Claudia Gonzalez-Espinosa
- Department of Pharmacobiology, Centro de Investigación y de Estudios Avanzados (Cinvestav), Unidad Sede Sur, Mexico City, Mexico
| | - Eva Salinas
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| |
Collapse
|
38
|
Fischer V, Haffner-Luntzer M. Interaction between bone and immune cells: Implications for postmenopausal osteoporosis. Semin Cell Dev Biol 2021; 123:14-21. [PMID: 34024716 DOI: 10.1016/j.semcdb.2021.05.014] [Citation(s) in RCA: 329] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Postmenopausal osteoporosis is a systemic disease characterized by the loss of bone mass and increased bone fracture risk largely resulting from significantly reduced levels of the hormone estrogen after menopause. Besides the direct negative effects of estrogen-deficiency on bone, indirect effects of altered immune status in postmenopausal women might contribute to ongoing bone destruction, as postmenopausal women often display a chronic low-grade inflammatory phenotype with altered cytokine expression and immune cell profile. In this context, it was previously shown that various immune cells interact with osteoblasts and osteoclasts either via direct cell-cell contact, or more likely via paracrine mechanisms. For example, specific subtypes of T lymphocytes express TNFα, which was shown to increase osteoblast apoptosis and to indirectly stimulate osteoclastogenesis via B cell-produced receptor-activator of NF-κB ligand (RANKL), thereby triggering bone loss during postmenopausal osteoporosis. Th17 cells release interleukin-17 (IL-17), which directs mesenchymal stem cell differentiation towards the osteogenic lineage, but also indirectly increases osteoclast differentiation. B lymphocytes are a major regulator of osteoclast formation via granulocyte colony-stimulating factor secretion and the RANKL/osteoprotegerin system under estrogen-deficient conditions. Macrophages might act differently on bone cells dependent on their polarization profile and their secreted paracrine factors, which might have implications for the development of postmenopausal osteoporosis, because macrophage polarization is altered during disease progression. Likewise, neutrophils play an important role during bone homeostasis, but their over-activation under estrogen-deficient conditions contributes to osteoblast apoptosis via the release of reactive oxygen species and increased osteoclastogenesis via RANKL signaling. Furthermore, mast cells might be involved in the development of postmenopausal osteoporosis, because they store high levels of osteoclastic mediators, including IL-6 and RANKL, in their granules and their numbers are greatly increased in osteoporotic bone. Additionally, bone fracture healing is altered under estrogen-deficient conditions with the increased presence of pro-inflammatory cytokines, including IL-6 and Midkine, which might contribute to healing disturbances. Consequently, in addition to the direct negative influence of estrogen-deficiency on bone, immune cell alterations contribute to the pathogenesis of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Verena Fischer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstraße 14, 89081 Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstraße 14, 89081 Ulm, Germany.
| |
Collapse
|
39
|
Luntzer K, Lackner I, Weber B, Mödinger Y, Ignatius A, Gebhard F, Mihaljevic SY, Haffner-Luntzer M, Kalbitz M. Increased Presence of Complement Factors and Mast Cells in Alveolar Bone and Tooth Resorption. Int J Mol Sci 2021; 22:ijms22052759. [PMID: 33803323 PMCID: PMC7967164 DOI: 10.3390/ijms22052759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/24/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Periodontitis is the inflammatory destruction of the tooth-surrounding and -supporting tissue, resulting at worst in tooth loss. Another locally aggressive disease of the oral cavity is tooth resorption (TR). This is associated with the destruction of the dental mineralized tissue. However, the underlying pathomechanisms remain unknown. The complement system, as well as mast cells (MCs), are known to be involved in osteoclastogenesis and bone loss. The complement factors C3 and C5 were previously identified as key players in periodontal disease. Therefore, we hypothesize that complement factors and MCs might play a role in alveolar bone and tooth resorption. To investigate this, we used the cat as a model because of the naturally occurring high prevalence of both these disorders in this species. Teeth, gingiva samples and serum were collected from domestic cats, which had an appointment for dental treatment under anesthesia, as well as from healthy cats. Histological analyses, immunohistochemical staining and the CH-50 and AH-50 assays revealed increased numbers of osteoclasts and MCs, as well as complement activity in cats with TR. Calcifications score in the gingiva was highest in animals that suffer from TR. This indicates that MCs and the complement system are involved in the destruction of the mineralized tissue in this condition.
Collapse
Affiliation(s)
- Kathrin Luntzer
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center, 89081 Ulm, Germany
- Small Animal Clinic Ravensburg Evidensia GmbH, Eywiesenstraße 4, 88212 Ravensburg, Germany
| | - Ina Lackner
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center, 89081 Ulm, Germany
| | - Birte Weber
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center, 89081 Ulm, Germany
| | - Yvonne Mödinger
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Institute of Orthopedic Research and Biomechanics, University of Ulm, 89081 Ulm, Germany
| | - Anita Ignatius
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Institute of Orthopedic Research and Biomechanics, University of Ulm, 89081 Ulm, Germany
| | - Florian Gebhard
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center, 89081 Ulm, Germany
| | | | - Melanie Haffner-Luntzer
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Institute of Orthopedic Research and Biomechanics, University of Ulm, 89081 Ulm, Germany
| | - Miriam Kalbitz
- Center for Trauma Research Ulm (ZTF), University of Ulm, 89081 Ulm, Germany; (K.L.); (I.L.); (B.W.); (Y.M.); (A.I.); (F.G.); (M.H.-L.)
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center, 89081 Ulm, Germany
- Correspondence:
| |
Collapse
|
40
|
Hao Z, Li J, Li B, Alder KD, Cahill SV, Munger AM, Lee I, Kwon HK, Back J, Xu S, Kang MJ, Lee FY. Smoking Alters Inflammation and Skeletal Stem and Progenitor Cell Activity During Fracture Healing in Different Murine Strains. J Bone Miner Res 2021; 36:186-198. [PMID: 32866293 PMCID: PMC9057220 DOI: 10.1002/jbmr.4175] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/16/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022]
Abstract
Smokers are at a higher risk of delayed union or nonunion after fracture repair. Few specific interventions are available for prevention because the molecular mechanisms that result in these negative sequelae are poorly understood. Murine models that mimic fracture healing in smokers are crucial in further understanding the local cellular and molecular alterations during fracture healing caused by smoking. We exposed three murine strains, C57BL/6J, 129X1/SvJ, and BALB/cJ, to cigarette smoke for 3 months before the induction of a midshaft transverse femoral osteotomy. We evaluated fracture healing 4 weeks after the osteotomy using radiography, micro-computed tomography (μCT), and biomechanical testing. Radiographic analysis demonstrated a significant decrease in the fracture healing capacity of smoking 129X1/SvJ mice. μCT results showed delayed remodeling of fracture calluses in all three strains after cigarette smoke exposure. Biomechanical testing indicated the most significant impairment in the functional properties of 129X1/SvJ in comparison with C57BL/6J and BALB/cJ mice after cigarette smoke exposure. Thus, the 129X1/SvJ strain is most suitable in simulating smoking-induced impaired fracture healing. Furthermore, in smoking 129X1/SvJ murine models, we investigated the molecular and cellular alterations in fracture healing caused by cigarette smoking using histology, flow cytometry, and multiplex cytokine/chemokine analysis. Histological analysis showed impaired chondrogenesis in cigarette smoking. In addition, the important reparative cell populations, including skeletal stem cells and their downstream progenitors, demonstrated decreased expansion after injury as a result of cigarette smoking. Moreover, significantly increased pro-inflammatory mediators and the recruitment of immune cells in fracture hematomas were demonstrated in smoking mice. Collectively, our findings demonstrate the significant cellular and molecular alterations during fracture healing impaired by smoking, including disrupted chondrogenesis, aberrant skeletal stem and progenitor cell activity, and a pronounced initial inflammatory response. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Zichen Hao
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Emergency and Trauma, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jun Li
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Bo Li
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Orthopaedics, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Kareme D Alder
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Sean V Cahill
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Alana M Munger
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Inkyu Lee
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - JungHo Back
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Shuogui Xu
- Department of Emergency and Trauma, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Min-Jong Kang
- Department of Medicine, Pulmonary, Critical Care, and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| |
Collapse
|
41
|
Mast Cell Functions Linking Innate Sensing to Adaptive Immunity. Cells 2020; 9:cells9122538. [PMID: 33255519 PMCID: PMC7761480 DOI: 10.3390/cells9122538] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Although mast cells (MCs) are known as key drivers of type I allergic reactions, there is increasing evidence for their critical role in host defense. MCs not only play an important role in initiating innate immune responses, but also influence the onset, kinetics, and amplitude of the adaptive arm of immunity or fine-tune the mode of the adaptive reaction. Intriguingly, MCs have been shown to affect T-cell activation by direct interaction or indirectly, by modifying the properties of antigen-presenting cells, and can even modulate lymph node-borne adaptive responses remotely from the periphery. In this review, we provide a summary of recent findings that explain how MCs act as a link between the innate and adaptive immunity, all the way from sensing inflammatory insult to orchestrating the final outcome of the immune response.
Collapse
|
42
|
Kundu D, Kennedy L, Meadows V, Baiocchi L, Alpini G, Francis H. The Dynamic Interplay Between Mast Cells, Aging/Cellular Senescence, and Liver Disease. Gene Expr 2020; 20:77-88. [PMID: 32727636 PMCID: PMC7650013 DOI: 10.3727/105221620x15960509906371] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mast cells are key players in acute immune responses that are evidenced by degranulation leading to a heightened allergic response. Activation of mast cells can trigger a number of different pathways contributing to metabolic conditions and disease progression. Aging results in irreversible physiological changes affecting all organs, including the liver. The liver undergoes senescence, changes in protein expression, and cell signaling phenotypes during aging, which regulate disease progression. Cellular senescence contributes to the age-related changes. Unsurprisingly, mast cells also undergo age-related changes in number, localization, and activation throughout their lifetime, which adversely affects the etiology and progression of many physiological conditions including liver diseases. In this review, we discuss the role of mast cells during aging, including features of aging (e.g., senescence) in the context of biliary diseases such as primary biliary cholangitis and primary sclerosing cholangitis and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Debjyoti Kundu
- *Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lindsey Kennedy
- *Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Vik Meadows
- *Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leonardo Baiocchi
- †Department of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Gianfranco Alpini
- *Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- ‡Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Heather Francis
- *Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- ‡Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
43
|
Muire PJ, Mangum LH, Wenke JC. Time Course of Immune Response and Immunomodulation During Normal and Delayed Healing of Musculoskeletal Wounds. Front Immunol 2020; 11:1056. [PMID: 32582170 PMCID: PMC7287024 DOI: 10.3389/fimmu.2020.01056] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/01/2020] [Indexed: 12/20/2022] Open
Abstract
Single trauma injuries or isolated fractures are often manageable and generally heal without complications. In contrast, high-energy trauma results in multi/poly-trauma injury patterns presenting imbalanced pro- and anti- inflammatory responses often leading to immune dysfunction. These injuries often exhibit delayed healing, leading to fibrosis of injury sites and delayed healing of fractures depending on the intensity of the compounding traumas. Immune dysfunction is accompanied by a temporal shift in the innate and adaptive immune cells distribution, triggered by the overwhelming release of an arsenal of inflammatory mediators such as complements, cytokines and damage associated molecular patterns (DAMPs) from necrotic cells. Recent studies have implicated this dysregulated inflammation in the poor prognosis of polytraumatic injuries, however, interventions focusing on immunomodulating inflammatory cellular composition and activation, if administered incorrectly, can result in immune suppression and unintended outcomes. Immunomodulation therapy is promising but should be conducted with consideration for the spatial and temporal distribution of the immune cells during impaired healing. This review describes the current state of knowledge in the spatiotemporal distribution patterns of immune cells at various stages during musculoskeletal wound healing, with a focus on recent advances in the field of Osteoimmunology, a study of the interface between the immune and skeletal systems, in long bone fractures. The goals of this review are to (1) discuss wound and fracture healing processes of normal and delayed healing in skeletal muscles and long bones; (2) provide a balanced perspective on temporal distributions of immune cells and skeletal cells during healing; and (3) highlight recent therapeutic interventions used to improve fracture healing. This review is intended to promote an understanding of the importance of inflammation during normal and delayed wound and fracture healing. Knowledge gained will be instrumental in developing novel immunomodulatory approaches for impaired healing.
Collapse
Affiliation(s)
- Preeti J. Muire
- Orthopaedic Trauma Research Department, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | | | | |
Collapse
|
44
|
Xie Y, Hu C, Feng Y, Li D, Ai T, Huang Y, Chen X, Huang L, Tan J. Osteoimmunomodulatory effects of biomaterial modification strategies on macrophage polarization and bone regeneration. Regen Biomater 2020; 7:233-245. [PMID: 32523726 PMCID: PMC7266668 DOI: 10.1093/rb/rbaa006] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/02/2020] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
Biomaterials as bone substitutes are always considered as foreign bodies that can trigger host immune responses. Traditional designing principles have been always aimed at minimizing the immune reactions by fabricating inert biomaterials. However, clinical evidence revealed that those methods still have limitations and many of which were only feasible in the laboratory. Currently, osteoimmunology, the very pioneering concept is drawing more and more attention-it does not simply regard the immune response as an obstacle during bone healing but emphasizes the intimate relationship of the immune and skeletal system, which includes diverse cells, cytokines, and signaling pathways. Properties of biomaterials like topography, wettability, surface charge, the release of cytokines, mediators, ions and other bioactive molecules can impose effects on immune responses to interfere with the skeletal system. Based on the bone formation mechanisms, the designing methods of the biomaterials change from immune evasive to immune reprogramming. Here, we discuss the osteoimmunomodulatory effects of the new modification strategies-adjusting properties of bone biomaterials to induce a favorable osteoimmune environment. Such strategies showed potential to benefit the development of bone materials and lay a solid foundation for the future clinical application.
Collapse
Affiliation(s)
- Yajuan Xie
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Cheng Hu
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Yi Feng
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Danfeng Li
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Tingting Ai
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Yulei Huang
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Xiaodan Chen
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Lijia Huang
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Jiali Tan
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| |
Collapse
|
45
|
Ignatius A, Sobacchi C. Editorial: Innate Immunity in the Context of Osteoimmunology. Front Immunol 2020; 11:603. [PMID: 32318075 PMCID: PMC7154185 DOI: 10.3389/fimmu.2020.00603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Affiliation(s)
- Anita Ignatius
- Medical Center, Institute of Orthopedic Research and Biomechanics, Ulm University, Ulm, Germany
| | - Cristina Sobacchi
- Milan Unit, CNR-IRGB, Milan, Italy.,Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| |
Collapse
|
46
|
Ragipoglu D, Dudeck A, Haffner-Luntzer M, Voss M, Kroner J, Ignatius A, Fischer V. The Role of Mast Cells in Bone Metabolism and Bone Disorders. Front Immunol 2020; 11:163. [PMID: 32117297 PMCID: PMC7025484 DOI: 10.3389/fimmu.2020.00163] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 01/21/2020] [Indexed: 12/15/2022] Open
Abstract
Mast cells (MCs) are important sensor and effector cells of the immune system that are involved in many physiological and pathological conditions. Increasing evidence suggests that they also play an important role in bone metabolism and bone disorders. MCs are located in the bone marrow and secrete a wide spectrum of mediators, which can be rapidly released upon activation of mature MCs following their differentiation in mucosal or connective tissues. Many of these mediators can exert osteocatabolic effects by promoting osteoclast formation [e.g., histamine, tumor necrosis factor (TNF), interleukin-6 (IL-6)] and/or by inhibiting osteoblast activity (e.g., IL-1, TNF). By contrast, MCs could potentially act in an osteoprotective manner by stimulating osteoblasts (e.g., transforming growth factor-β) or reducing osteoclastogenesis (e.g., IL-12, interferon-γ). Experimental studies investigating MC functions in physiological bone turnover using MC-deficient mouse lines give contradictory results, reporting delayed or increased bone turnover or no influence depending on the mouse model used. By contrast, the involvement of MCs in various pathological conditions affecting bone is evident. MCs may contribute to the pathogenesis of primary and secondary osteoporosis as well as inflammatory disorders, including rheumatoid arthritis and osteoarthritis, because increased numbers of MCs were found in patients suffering from these diseases. The clinical observations could be largely confirmed in experimental studies using MC-deficient mouse models, which also provide mechanistic insights. MCs also regulate bone healing after fracture by influencing the inflammatory response toward the fracture, vascularization, bone formation, and callus remodeling by osteoclasts. This review summarizes the current view and understanding of the role of MCs on bone in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Deniz Ragipoglu
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anne Dudeck
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Melanie Haffner-Luntzer
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Martin Voss
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Jochen Kroner
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anita Ignatius
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Verena Fischer
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
47
|
Guder C, Gravius S, Burger C, Wirtz DC, Schildberg FA. Osteoimmunology: A Current Update of the Interplay Between Bone and the Immune System. Front Immunol 2020; 11:58. [PMID: 32082321 PMCID: PMC7004969 DOI: 10.3389/fimmu.2020.00058] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Immunology, already a discipline in its own right, has become a major part of many different medical fields. However, its relationship to orthopedics and trauma surgery has unfortunately, and perhaps unjustly, been developing rather slowly. Discoveries in recent years have emphasized the immense breadth of communication and connection between both systems and, importantly, the highly promising therapeutic opportunities. Recent discoveries of factors originally assigned to the immune system have now also been shown to have a significant impact on bone health and disease, which has greatly changed how we approach treatment of bone pathologies. In case of bone fracture, immune cells, especially macrophages, are present throughout the whole healing process, assure defense against pathogens and discharge a complex variety of effectors to regulate bone modeling. In rheumatoid arthritis and osteoporosis, the immune system contributes to the formation of the pathological and chronic conditions. Fascinatingly, prosthesis failure is not at all solely a mechanical problem of improper strain but works in conjunction with an active contribution of the immune system as a reaction to irritant debris from material wear. Unraveling conjoined mechanisms of the immune and osseous systems heralds therapeutic possibilities for ailments of both. Contemplation of the bone as merely an unchanging support pillar is outdated and obsolete. Instead it is mandatory that this highly diverse network be incorporated in our understanding of the immune system and hematopoiesis.
Collapse
Affiliation(s)
- Christian Guder
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Sascha Gravius
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany.,Department of Orthopedics and Trauma Surgery, University Medical Center Mannheim of University Heidelberg, Mannheim, Germany
| | - Christof Burger
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C Wirtz
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
48
|
Min HK, Kim KW, Lee SH, Kim HR. Roles of mast cells in rheumatoid arthritis. Korean J Intern Med 2020; 35:12-24. [PMID: 31722515 PMCID: PMC6960056 DOI: 10.3904/kjim.2019.271] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory arthritis, and the complex interaction and activation of innate and adaptive immune cells are involved in RA pathogenesis. Mast cells (MCs) are one of the tissue-resident innate immune cells, and they contribute to RA pathogenesis. In the present review, the evidence of the pathologic role of MC in RA is discussed based on human and animal data. In addition, the potential role of MC in RA pathogenesis and the research area that should be focused on in the future are suggested.
Collapse
Affiliation(s)
- Hong Ki Min
- Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, Seoul, Korea
| | - Kyoung-Woon Kim
- Conversant Research Consortium in Immunologic Disease, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sang-Heon Lee
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
| | - Hae-Rim Kim
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
- Correspondence to Hae-Rim Kim, M.D. Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Korea Tel: +82-2-2030-7542, Fax: +82-2-2030-7728, E-mail:
| |
Collapse
|
49
|
Gruber R. Osteoimmunology: Inflammatory osteolysis and regeneration of the alveolar bone. J Clin Periodontol 2019; 46 Suppl 21:52-69. [PMID: 30623453 DOI: 10.1111/jcpe.13056] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/09/2018] [Accepted: 12/06/2018] [Indexed: 02/06/2023]
Abstract
AIM Osteoimmunology covers the cellular and molecular mechanisms responsible for inflammatory osteolysis that culminates in the degradation of alveolar bone. Osteoimmunology also focuses on the interplay of immune cells with bone cells during bone remodelling and regeneration. The aim of this review was to provide insights into how osteoimmunology affects alveolar bone health and disease. METHOD This review is based on a narrative approach to assemble mouse models that provide insights into the cellular and molecular mechanisms causing inflammatory osteolysis and on the impact of immune cells on alveolar bone regeneration. RESULTS Mouse models have revealed the molecular pathways by which microbial and other factors activate immune cells that initiate an inflammatory response. The inflammation-induced alveolar bone loss occurs with the concomitant suppression of bone formation. Mouse models also showed that immune cells contribute to the resolution of inflammation and bone regeneration, even though studies with a focus on alveolar socket healing are rare. CONCLUSIONS Considering that osteoimmunology is evolutionarily conserved, osteolysis removes the cause of inflammation by provoking tooth loss. The impact of immune cells on bone regeneration is presumably a way to reinitiate the developmental mechanisms of intramembranous and endochondral bone formation.
Collapse
Affiliation(s)
- Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, Vienna, Austria.,Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
50
|
Bone Anabolic Response in the Calvaria Following Mild Traumatic Brain Injury is Mediated by the Cannabinoid-1 Receptor. Sci Rep 2019; 9:16196. [PMID: 31700010 PMCID: PMC6838196 DOI: 10.1038/s41598-019-51720-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/27/2019] [Indexed: 12/28/2022] Open
Abstract
Brain trauma was clinically associated with increased osteogenesis in the appendicular skeleton. We showed previously in C57BL/6J mice that mild traumatic brain injury (mTBI) transiently induced bone formation in the femur via the cannabinoid-1 (CB1) receptor. Here, we subjected ICR mice to mTBI and examined the bone response in the skull using microCT. We also measured mast cell degranulation (MCD)72 h post-injury. Finally, we measured brain and calvarial endocannabinoids levels post-mTBI. mTBI led to decreased bone porosity on the contralateral (untouched) side. This effect was apparent both in young and mature mice. Administration of rimonabant (CB1 inverse agonist) completely abrogated the effect of mTBI on calvarial porosity and significantly reduced MCD, compared with vehicle-treated controls. We also found that mTBI resulted in elevated levels of anandamide, but not 2-arachidonoylglycerol, in the contralateral calvarial bone, whereas brain levels remained unchanged. In C57BL/6J CB1 knockout mice, mTBI did not reduce porosity but in general the porosity was significantly lower than in WT controls. Our findings suggest that mTBI induces a strain-specific CB1-dependent bone anabolic response in the skull, probably mediated by anandamide, but seemingly unrelated to inflammation. The endocannabinoid system is therefore a plausible target in management of bone response following head trauma.
Collapse
|