1
|
Mi B, Xiong Y, Knoedler S, Alfertshofer M, Panayi AC, Wang H, Lin S, Li G, Liu G. Ageing-related bone and immunity changes: insights into the complex interplay between the skeleton and the immune system. Bone Res 2024; 12:42. [PMID: 39103328 DOI: 10.1038/s41413-024-00346-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/16/2024] [Accepted: 05/27/2024] [Indexed: 08/07/2024] Open
Abstract
Ageing as a natural irreversible process inherently results in the functional deterioration of numerous organ systems and tissues, including the skeletal and immune systems. Recent studies have elucidated the intricate bidirectional interactions between these two systems. In this review, we provide a comprehensive synthesis of molecular mechanisms of cell ageing. We further discuss how age-related skeletal changes influence the immune system and the consequent impact of immune system alterations on the skeletal system. Finally, we highlight the clinical implications of these findings and propose potential strategies to promote healthy ageing and reduce pathologic deterioration of both the skeletal and immune systems.
Collapse
Affiliation(s)
- Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Samuel Knoedler
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Michael Alfertshofer
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig - Maximilian University Munich, Munich, Germany
| | - Adriana C Panayi
- Division of Plastic Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Hand-, Plastic and Reconstructive Surgery, Microsurgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Haixing Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China.
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China.
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
2
|
Song C, Liu Y, Tao X, Cheng K, Cai W, Zhou D, Zhou Y, Wang L, Shi H, Hao Q, Liu Z. Immunomodulation Pathogenesis and Treatment of Bone Nonunion. Orthop Surg 2024; 16:1770-1782. [PMID: 38946017 PMCID: PMC11293939 DOI: 10.1111/os.14131] [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: 02/05/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 07/02/2024] Open
Abstract
Fractures and bone nonunion commonly require surgical intervention. Serious outcomes of non-healing in the late stages of fracture place a significant financial burden on society and families. Bone nonunion occurs when a fracture stops healing, for many reasons, and leads to a variety of bad outcomes. Numerous factors, including biomechanics and immunology, are involved in the complicated mechanisms of bone nonunion. The immune-inflammatory response plays a significant part in the emergence of bone nonunion, and the occurrence, control, and remission of inflammation in the bone healing process have a significant influence on the ultimate success of bone tissue repair. In the bone microenvironment, immune cells and associated cytokines control bone repair, which is significantly influenced by macrophages, T cells, and fibroblast growth factor. To limit acute inflammation and balance osteogenesis and osteoblastogenesis for tissue repair and regeneration, immune cells and various cytokines in the local microenvironment must be precisely regulated. As a bad complication of late-stage fractures, bone nonunion has a significant effect on patients' quality of life and socioeconomic development. Therefore, in-depth research on its pathogenesis and treatment methods has important clinical value. To provide more precise, focused therapeutic options for the treatment of bone nonunion, we discuss the regulatory roles of the key immune cells engaged in bone healing within the microenvironment during bone healing and their effect on osteogenesis.
Collapse
Affiliation(s)
- Chao Song
- Department of Orthopedics and Traumatology (Trauma and Bone‐Setting), Laboratory of Integrated Chinese and Western Medicine for Orthopedic and Traumatic Diseases Prevention and Treatment, The Affiliated Traditional Chinese Medicine HospitalSouthwest Medical UniversityLuzhouChina
| | - Yong Liu
- Department of Bone and Joint Sports MedicineXingguo People's Hospital, Gannan Medical CollegeXingguoChina
| | - Xingxing Tao
- College of Integrative Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine HospitalSouthwest Medical UniversityLuzhouChina
| | - Kang Cheng
- Department of Orthopedics and Traumatology (Trauma and Bone‐Setting), Laboratory of Integrated Chinese and Western Medicine for Orthopedic and Traumatic Diseases Prevention and Treatment, The Affiliated Traditional Chinese Medicine HospitalSouthwest Medical UniversityLuzhouChina
| | - Weiye Cai
- Department of Orthopedics and Traumatology (Trauma and Bone‐Setting), Laboratory of Integrated Chinese and Western Medicine for Orthopedic and Traumatic Diseases Prevention and Treatment, The Affiliated Traditional Chinese Medicine HospitalSouthwest Medical UniversityLuzhouChina
| | - Daqian Zhou
- Department of Orthopedics and Traumatology (Trauma and Bone‐Setting), Laboratory of Integrated Chinese and Western Medicine for Orthopedic and Traumatic Diseases Prevention and Treatment, The Affiliated Traditional Chinese Medicine HospitalSouthwest Medical UniversityLuzhouChina
| | - Yang Zhou
- Department of Orthopedics and Traumatology (Trauma and Bone‐Setting), Laboratory of Integrated Chinese and Western Medicine for Orthopedic and Traumatic Diseases Prevention and Treatment, The Affiliated Traditional Chinese Medicine HospitalSouthwest Medical UniversityLuzhouChina
| | - Liquan Wang
- Department of Orthopedics and Traumatology (Trauma and Bone‐Setting), Laboratory of Integrated Chinese and Western Medicine for Orthopedic and Traumatic Diseases Prevention and Treatment, The Affiliated Traditional Chinese Medicine HospitalSouthwest Medical UniversityLuzhouChina
| | - Houyin Shi
- Department of Orthopedics and Traumatology (Trauma and Bone‐Setting), Laboratory of Integrated Chinese and Western Medicine for Orthopedic and Traumatic Diseases Prevention and Treatment, The Affiliated Traditional Chinese Medicine HospitalSouthwest Medical UniversityLuzhouChina
| | - Qi Hao
- Orthopedic Surgery, The Affiliated Traditional Chinese Medicine HospitalSouthwest Medical UniversityLuzhouChina
| | - Zongchao Liu
- Department of Orthopedics and Traumatology (Trauma and Bone‐Setting), Laboratory of Integrated Chinese and Western Medicine for Orthopedic and Traumatic Diseases Prevention and Treatment, The Affiliated Traditional Chinese Medicine HospitalSouthwest Medical UniversityLuzhouChina
- Department of OrthopedicsLuzhou Longmatan District People's HospitalLuzhouChina
| |
Collapse
|
3
|
Liu X, Li H. Global trends in research on aging associated with periodontitis from 2002 to 2023: a bibliometric analysis. Front Endocrinol (Lausanne) 2024; 15:1374027. [PMID: 38800469 PMCID: PMC11116588 DOI: 10.3389/fendo.2024.1374027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/02/2024] [Indexed: 05/29/2024] Open
Abstract
Background Aging has been implicated in many chronic inflammatory diseases, including periodontitis. Periodontitis is an inflammatory disease caused by long-term irritation of the periodontal tissues by the plaque biofilm on the surface of the teeth. However, only a few bibliometric analyses have systematically studied this field to date. This work sought to visualize research hot spots and trends in aging associated with periodontitis from 2002 to 2023 through bibliometric approaches. Methods Graphpad prism v8.0.2 was used to analyse and plot annual papers, national publication trends and national publication heat maps. In addition, CtieSpace (6.1.6R (64-bit) Advanced Edition) and VOSviewer (version 1.6.18) were used to analyse these data and visualize the scientific knowledge graph. Results The number of documents related to aging associated with periodontitis has steadily increased over 21 years. With six of the top ten institutions in terms of publications coming from the US, the US is a major driver of research in this area. journal of periodontology is the most published journal in the field. Tonetti MS is the most prolific authors and co-cited authors in the field. Journal of Periodontology and Journal of Clinical Periodontology are the most popular journals in the field with the largest literature. Periodontitis, Alzheimer's disease, and peri-implantitis are current hot topics and trends in the field. Inflammation, biomarkers, oxidative stress cytokines are current research hotspots in this field. Conclusion Our research found that global publications regarding research on aging associated with periodontitis increased dramatically and were expected to continue increasing. Inflammation and aging, and the relationship between periodontitis and systemic diseases, are topics worthy of attention.
Collapse
Affiliation(s)
| | - Hongjiao Li
- Department of Stomatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Gou M, Wang H, Xie H, Song H. Macrophages in guided bone regeneration: potential roles and future directions. Front Immunol 2024; 15:1396759. [PMID: 38736888 PMCID: PMC11082316 DOI: 10.3389/fimmu.2024.1396759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Guided bone regeneration (GBR) is one of the most widely used and thoroughly documented alveolar bone augmentation surgeries. However, implanting GBR membranes inevitably triggers an immune response, which can lead to inflammation and failure of bone augmentation. It has been shown that GBR membranes may significantly improve in vivo outcomes as potent immunomodulators, rather than solely serving as traditional barriers. Macrophages play crucial roles in immune responses and participate in the entire process of bone injury repair. The significant diversity and high plasticity of macrophages complicate our understanding of the immunomodulatory mechanisms underlying GBR. This review provides a comprehensive summary of recent findings on the potential role of macrophages in GBR for bone defects in situ. Specifically, macrophages can promote osteogenesis or fibrous tissue formation in bone defects and degradation or fibrous encapsulation of membranes. Moreover, GBR membranes can influence the recruitment and polarization of macrophages. Therefore, immunomodulating GBR membranes are primarily developed by improving macrophage recruitment and aggregation as well as regulating macrophage polarization. However, certain challenges remain to be addressed in the future. For example, developing more rational and sophisticated sequential delivery systems for macrophage activation reagents; addressing the interference of bone graft materials and dental implants; and understanding the correlations among membrane degradation, macrophage responses, and bone regeneration.
Collapse
Affiliation(s)
- Min Gou
- Department of Stomatology, Chengdu Second People’s Hospital, Chengdu, China
| | - Hang Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Huiqi Xie
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Hongjie Song
- Department of Stomatology, Chengdu Second People’s Hospital, Chengdu, China
| |
Collapse
|
5
|
Stevens HY, Jimenez AC, Wang B, Li Y, Selvam S, Bowles-Welch AC. Mesenchymal Stromal Cell (MSC) Functional Analysis-Macrophage Activation and Polarization Assays. Bio Protoc 2024; 14:e4957. [PMID: 38841292 PMCID: PMC10958173 DOI: 10.21769/bioprotoc.4957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 06/07/2024] Open
Abstract
Stem cell-based therapies have evolved to become a key component of regenerative medicine approaches to human pathologies. Exogenous stem cell transplantation takes advantage of the potential of stem cells to self-renew, differentiate, home to sites of injury, and sufficiently evade the immune system to remain viable for the release of anti-inflammatory cytokines, chemokines, and growth factors. Common to many pathologies is the exacerbation of inflammation at the injury site by proinflammatory macrophages. An increasing body of evidence has demonstrated that mesenchymal stromal cells (MSCs) can influence the immunophenotype and function of myeloid lineage cells to promote therapeutic effects. Understanding the degree to which MSCs can modulate the phenotype of macrophages within an inflammatory environment is of interest when considering strategies for targeted cell therapies. There is a critical need for potency assays to elucidate these intercellular interactions in vitro and provide insight into potential mechanisms of action attributable to the immunomodulatory and polarizing capacities of MSCs, as well as other cells with immunomodulatory potential. However, the complexity of the responses, in terms of cell phenotypes and characteristics, timing of these interactions, and the degree to which cell contact is involved, have made the study of these interactions challenging. To provide a research tool to study the direct interactions between MSCs and macrophages, we developed a potency assay that directly co-cultures MSCs with naïve macrophages under proinflammatory conditions. Using this assay, we demonstrated changes in the macrophage secretome and phenotype, which can be used to evaluate the abilities of the cell samples to influence the cell microenvironment. These results suggest the immunomodulatory effects of MSCs on macrophages while revealing key cytokines and phenotypic changes that may inform their efficacy as potential cellular therapies. Key features • The protocol uses monocytes differentiated into naïve macrophages, which are loosely adherent, have a relatively homogeneous genetic background, and resemble peripheral blood mononuclear cells-derived macrophages. • The protocol requires a plate reader and a flow cytometer with the ability to detect six fluorophores. • The protocol provides a quantitative measurement of co-culture conditions by the addition of a fixed number of freshly thawed or culture-rescued MSCs to macrophages. • This protocol uses assessment of the secretome and cell harvest to independently verify the nature of the interactions between macrophages and MSCs.
Collapse
Affiliation(s)
- Hazel Y. Stevens
- Marcus Center for Therapeutic Cell Characterization
and Manufacturing, Institute for Bioengineering and Bioscience, Georgia
Institute of Technology, Atlanta, GA, USA
| | - Angela C. Jimenez
- Marcus Center for Therapeutic Cell Characterization
and Manufacturing, Institute for Bioengineering and Bioscience, Georgia
Institute of Technology, Atlanta, GA, USA
- The Wallace H. Coulter Department of Biomedical
Engineering, Georgia Tech and Emory University, Atlanta, GA, USA
- The National Science Foundation (NSF) Engineering
Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia
Institute of Technology, Atlanta, GA, USA
| | - Bryan Wang
- Marcus Center for Therapeutic Cell Characterization
and Manufacturing, Institute for Bioengineering and Bioscience, Georgia
Institute of Technology, Atlanta, GA, USA
- The Wallace H. Coulter Department of Biomedical
Engineering, Georgia Tech and Emory University, Atlanta, GA, USA
- The National Science Foundation (NSF) Engineering
Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia
Institute of Technology, Atlanta, GA, USA
| | - Ye Li
- Marcus Center for Therapeutic Cell Characterization
and Manufacturing, Institute for Bioengineering and Bioscience, Georgia
Institute of Technology, Atlanta, GA, USA
| | - Shivaram Selvam
- Marcus Center for Therapeutic Cell Characterization
and Manufacturing, Institute for Bioengineering and Bioscience, Georgia
Institute of Technology, Atlanta, GA, USA
- The National Science Foundation (NSF) Engineering
Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia
Institute of Technology, Atlanta, GA, USA
| | - Annie C. Bowles-Welch
- Marcus Center for Therapeutic Cell Characterization
and Manufacturing, Institute for Bioengineering and Bioscience, Georgia
Institute of Technology, Atlanta, GA, USA
- The National Science Foundation (NSF) Engineering
Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Georgia
Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
6
|
Zhao H, Zhang W, Liu Y, Peng W. Risk factors and prediction model for thrombocytopenia following coronary artery bypass graft surgery in elderly Chinese population. J Thorac Dis 2024; 16:273-284. [PMID: 38410552 PMCID: PMC10894372 DOI: 10.21037/jtd-23-1396] [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: 09/05/2023] [Accepted: 12/01/2023] [Indexed: 02/28/2024]
Abstract
Background Thrombocytopenia, a common complication of coronary artery bypass graft (CABG) surgery, is particularly prevalent among elderly individuals. This study developed a risk prediction model utilizing preoperative and intraoperative variables to identify high-risk elderly patients prone to developing thrombocytopenia. Methods The patients were retrospectively recruited from Beijing Anzhen Hospital between February 2019 and December 2020. Postoperative thrombocytopenia was defined as a postoperative platelet (PLT) count <100×109/L as measured within 7 days after surgery. The entire population was randomly split into derivation and validation sets in a 7:3 ratio. The derivation set underwent variable screen by the least absolute shrinkage and selection operator (LASSO) regression method. To evaluate the predictive ability of the model for thrombocytopenia, decision curve analysis (DCA) and receiver operating characteristic (ROC) curves were generated in the derivation and validation sets. Results A total of 1,773 patients were recruited in this study, with random assignment to either the derivation set (1,242 cases) or the validation set (531 cases). LASSO regression was utilized the risk factors associated with thrombocytopenia, resulting in selection of preoperative baseline variables: body mass index (BMI), estimated glomerular filtration rate (eGFR), B-type natriuretic peptide (BNP), preoperative PLT, and use of beta-blocker, and intraoperative variables: red blood cell (RBC) transfusion, plasma transfusion, use of intra-aortic balloon pump (IABP) and cardiopulmonary bypass (CPB), reoperation for bleeding, washed RBC transfusion volume, and use of epinephrine. The logistic regression was employed to establish the risk prediction. The area under the ROC curve (AUC) for the derivation set was 0.900 [95% confidence interval (CI): 0.880-0.920], while for the validation cohort, it was 0.897 (95% CI: 0.866-0.928). Conclusions The model incorporating significant preoperative and intraoperative variables exhibited good predictive performance for thrombocytopenia in elderly patients undergoing CABG surgery.
Collapse
Affiliation(s)
- Honglei Zhao
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Wei Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Yongmin Liu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Wenxing Peng
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Minucci SB, Heise RL, Reynolds AM. Agent-based vs. equation-based multi-scale modeling for macrophage polarization. PLoS One 2024; 19:e0270779. [PMID: 38271449 PMCID: PMC10810539 DOI: 10.1371/journal.pone.0270779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/29/2023] [Indexed: 01/27/2024] Open
Abstract
Macrophages show high plasticity and result in heterogenic subpopulations or polarized states identified by specific cellular markers. These immune cells are typically characterized as pro-inflammatory, or classically activated M1, and anti-inflammatory, or alternatively activated M2. However, a more precise definition places them along a spectrum of activation where they may exhibit a number of pro- or anti-inflammatory roles. To understand M1-M2 dynamics in the context of a localized response and explore the results of different mathematical modeling approaches based on the same biology, we utilized two different modeling techniques, ordinary differential equation (ODE) modeling and agent-based modeling (ABM), to simulate the spectrum of macrophage activation to general pro- and anti-inflammatory stimuli on an individual and multi-cell level. The ODE model includes two hallmark pro- and anti-inflammatory signaling pathways and the ABM incorporates similar M1-M2 dynamics but in a spatio-temporal platform. Both models link molecular signaling with cellular-level dynamics. We then performed simulations with various initial conditions to replicate different experimental setups. Similar results were observed in both models after tuning to a common calibrating experiment. Comparing the two models' results sheds light on the important features of each modeling approach. When more data is available these features can be considered when choosing techniques to best fit the needs of the modeler and application.
Collapse
Affiliation(s)
- Sarah B. Minucci
- Department of Mathematics & Applied Mathematics, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Rebecca L. Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Angela M. Reynolds
- Department of Mathematics & Applied Mathematics, Virginia Commonwealth University, Richmond, VA, United States of America
| |
Collapse
|
8
|
Zhang L, Guan Q, Wang Z, Feng J, Zou J, Gao B. Consequences of Aging on Bone. Aging Dis 2023:AD.2023.1115. [PMID: 38029404 DOI: 10.14336/ad.2023.1115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023] Open
Abstract
With the aging of the global population, the incidence of musculoskeletal diseases has been increasing, seriously affecting people's health. As people age, the microenvironment within skeleton favors bone resorption and inhibits bone formation, accompanied by bone marrow fat accumulation and multiple cellular senescence. Specifically, skeletal stem/stromal cells (SSCs) during aging tend to undergo adipogenesis rather than osteogenesis. Meanwhile, osteoblasts, as well as osteocytes, showed increased apoptosis, decreased quantity, and multiple functional limitations including impaired mechanical sensing, intercellular modulation, and exosome secretion. Also, the bone resorption function of macrophage-lineage cells (including osteoclasts and preosteoclasts) was significantly enhanced, as well as impaired vascularization and innervation. In this study, we systematically reviewed the effect of aging on bone and the within microenvironment (including skeletal cells as well as their intracellular structure variations, vascular structures, innervation, marrow fat distribution, and lymphatic system) caused by aging, and mechanisms of osteoimmune regulation of the bone environment in the aging state, and the causal relationship with multiple musculoskeletal diseases in addition with their potential therapeutic strategy.
Collapse
Affiliation(s)
- Lingli Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai, China
| | - Qiao Guan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Zhikun Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Jie Feng
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Jun Zou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Bo Gao
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
9
|
Lauzier DC, Chiang SN, Moran CJ. Etiologies of Brain Arteriovenous Malformation Recurrence: A Focus on Pediatric Disease. Pediatr Neurol 2023; 148:94-100. [PMID: 37690270 DOI: 10.1016/j.pediatrneurol.2023.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/18/2023] [Accepted: 08/15/2023] [Indexed: 09/12/2023]
Abstract
Pediatric brain arteriovenous malformations are a major cause of morbidity and mortality, with the harmful effects of this disease compounded by the additional disability-years experienced by children with ruptured or other symptomatic arteriovenous malformations. In addition to the risks shared with their adult counterparts, pediatric patients frequently experience recurrence following radiographic cure, which presents an additional source of morbidity and mortality. Therefore, there is a need to synthesize potential mechanisms contributing to the elevated recurrence risk in the pediatric population and discuss how these translate to practical considerations for managing these patients.
Collapse
Affiliation(s)
- David C Lauzier
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri.
| | - Sarah N Chiang
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Christopher J Moran
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
10
|
Riegger J, Schoppa A, Ruths L, Haffner-Luntzer M, Ignatius A. Oxidative stress as a key modulator of cell fate decision in osteoarthritis and osteoporosis: a narrative review. Cell Mol Biol Lett 2023; 28:76. [PMID: 37777764 PMCID: PMC10541721 DOI: 10.1186/s11658-023-00489-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/11/2023] [Indexed: 10/02/2023] Open
Abstract
During aging and after traumatic injuries, cartilage and bone cells are exposed to various pathophysiologic mediators, including reactive oxygen species (ROS), damage-associated molecular patterns, and proinflammatory cytokines. This detrimental environment triggers cellular stress and subsequent dysfunction, which not only contributes to the development of associated diseases, that is, osteoporosis and osteoarthritis, but also impairs regenerative processes. To counter ROS-mediated stress and reduce the overall tissue damage, cells possess diverse defense mechanisms. However, cellular antioxidative capacities are limited and thus ROS accumulation can lead to aberrant cell fate decisions, which have adverse effects on cartilage and bone homeostasis. In this narrative review, we address oxidative stress as a major driver of pathophysiologic processes in cartilage and bone, including senescence, misdirected differentiation, cell death, mitochondrial dysfunction, and impaired mitophagy by illustrating the consequences on tissue homeostasis and regeneration. Moreover, we elaborate cellular defense mechanisms, with a particular focus on oxidative stress response and mitophagy, and briefly discuss respective therapeutic strategies to improve cell and tissue protection.
Collapse
Affiliation(s)
- Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, 89081, Ulm, Germany.
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Leonie Ruths
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| |
Collapse
|
11
|
Fan S, Sun X, Su C, Xue Y, Song X, Deng R. Macrophages-bone marrow mesenchymal stem cells crosstalk in bone healing. Front Cell Dev Biol 2023; 11:1193765. [PMID: 37427382 PMCID: PMC10327485 DOI: 10.3389/fcell.2023.1193765] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023] Open
Abstract
Bone healing is associated with many orthopedic conditions, including fractures and osteonecrosis, arthritis, metabolic bone disease, tumors and periprosthetic particle-associated osteolysis. How to effectively promote bone healing has become a keen topic for researchers. The role of macrophages and bone marrow mesenchymal stem cells (BMSCs) in bone healing has gradually come to light with the development of the concept of osteoimmunity. Their interaction regulates the balance between inflammation and regeneration, and when the inflammatory response is over-excited, attenuated, or disturbed, it results in the failure of bone healing. Therefore, an in-depth understanding of the function of macrophages and bone marrow mesenchymal stem cells in bone regeneration and the relationship between the two could provide new directions to promote bone healing. This paper reviews the role of macrophages and bone marrow mesenchymal stem cells in bone healing and the mechanism and significance of their interaction. Several new therapeutic ideas for regulating the inflammatory response in bone healing by targeting macrophages and bone marrow mesenchymal stem cells crosstalk are also discussed.
Collapse
Affiliation(s)
- Siyu Fan
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xin Sun
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Chuanchao Su
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yiwen Xue
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xiao Song
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Runzhi Deng
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
12
|
Kushioka J, Toya M, Shen H, Hirata H, Zhang N, Huang E, Tsubosaka M, Gao Q, Teissier V, Li X, Utsunomiya T, Goodman SB. Therapeutic effects of MSCs, genetically modified MSCs, and NFĸB-inhibitor on chronic inflammatory osteolysis in aged mice. J Orthop Res 2023; 41:1004-1013. [PMID: 36031590 PMCID: PMC9971358 DOI: 10.1002/jor.25434] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 02/04/2023]
Abstract
The number of total joint replacements is increasing, especially in elderly patients, and so too are implant-related complications such as prosthesis loosening. Wear particles from the prosthesis induce a chronic inflammatory reaction and subsequent osteolysis, leading to the need for revision surgery. This study investigated the therapeutic effect of NF-ĸB decoy oligodeoxynucleotides (ODN), mesenchymal stem cells (MSCs), and genetically-modified NF-ĸB sensing interleukin-4 over-secreting MSCs (IL4-MSCs) on chronic inflammation in aged mice. The model was generated by continuous infusion of contaminated polyethylene particles into the intramedullary space of the distal femur of aged mice (15-17 months old) for 6 weeks. Local delivery of ODN showed increased bone mineral density (BMD), decreased osteoclast-like cells, increased alkaline phosphatase (ALP)-positive area, and increased M2/M1 macrophage ratio. Local injection of MSCs and IL4-MSCs significantly decreased osteoclast-like cells and increased the M2/M1 ratio, with a greater trend for IL4-MSCs than MSCs. MSCs significantly increased ALP-positive area and BMD values compared with the control. The IL4-MSCs demonstrated higher values for both ALP-positive area and BMD. These findings demonstrated the therapeutic effects of ODN, MSCs, and IL4-MSCs on chronic inflammatory osteolysis in aged mice. The two MSC-based therapies were more effective than ODN in increasing the M2/M1 macrophage ratio, reducing bone resorption, and increasing bone formation. Specifically, MSCs were more effective in increasing bone formation, and IL4-MSCs were more effective in mitigating inflammation. This study suggests potential therapeutic strategies for treating wear particle-associated inflammatory osteolysis after arthroplasty in the elderly.
Collapse
Affiliation(s)
- Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Hirohito Hirata
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Ejun Huang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Victoria Teissier
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Xueping Li
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | | | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
13
|
Valentine MS, Weigel C, Kamga Gninzeko F, Tho C, Gräler MH, Reynolds AM, Spiegel S, Heise RL. S1P lyase inhibition prevents lung injury following high pressure-controlled mechanical ventilation in aging mice. Exp Gerontol 2023; 173:112074. [PMID: 36566871 PMCID: PMC9975034 DOI: 10.1016/j.exger.2022.112074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Ventilator-induced Lung Injury (VILI) is characterized by hypoxia, inflammatory cytokine influx, loss of alveolar barrier integrity, and decreased lung compliance. Aging influences lung structure and function and is a predictive factor in the severity of VILI; however, the mechanisms of aging that influence the progression or increased susceptibility remain unknown. Aging impacts immune system function and may increase inflammation in healthy individuals. Recent studies suggest that the bioactive sphingolipid mediator sphingosine-1-phosphate (S1P) and the enzyme that degrades it S1P lyase (SPL) may be involved in lung pathologies including acute lung injury. It is unknown whether aging influences S1P and SPL expression that have been implicated in lung inflammation, injury, and cell apoptosis. We hypothesized that aging and injurious mechanical ventilation synergistically impair S1P levels and enhance S1P lyase (SPL) expression that amplifies alveolar barrier damage and diminishes pulmonary function. Young (2-3 mo) and old (20-25 mo) C57BL/6 mice were mechanically ventilated for 2 h using pressure-controlled mechanical ventilation (PCMV) at 45 cmH2O and 35 cmH2O, respectively. We assessed the impact of aging and PCMV on several indications of acute lung injury, immune cell recruitment, S1P levels and SPL activity. Furthermore, we evaluated the protective effects of inhibiting SPL by tetrahydroxybutylimidazol (THI) administration on the negative outcomes associated with aging and mechanical injury. PCMV exacerbated lung injury in old mice and increased neutrophil influx that was further exacerbated due to aging. SPL expression increased in the young and old ventilated mice and the old nonventilated group. THI treatment reduced several of the indicators of lung injury and resulted in elevated S1P levels in lung tissue and plasma from mice that were injured from mechanical ventilation. CD80 and CD206 activation markers of alveolar and interstitial macrophages were also influenced by THI. SPL inhibition may be a viable therapeutic approach for patients requiring mechanical ventilation by preventing or regulating the exaggerated inflammatory response and reducing lung injury.
Collapse
Affiliation(s)
- M S Valentine
- Department of Biomedical Engineering, Virginia Commonwealth University, United States of America
| | - C Weigel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, United States of America
| | - F Kamga Gninzeko
- Department of Biomedical Engineering, Virginia Commonwealth University, United States of America
| | - C Tho
- Department of Biomedical Engineering, Virginia Commonwealth University, United States of America
| | - M H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Center for Molecular Biomedicine (CMB) and Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - A M Reynolds
- Department of Mathematics and Applied Mathematics, Virginia Commonwealth University, United States of America
| | - S Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, United States of America
| | - R L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, United States of America.
| |
Collapse
|
14
|
Zoledronate/Anti-VEGF Neutralizing Antibody Combination Administration Increases Osteal Macrophages in a Murine Model of MRONJ Stage 0-like Lesions. J Clin Med 2023; 12:jcm12051914. [PMID: 36902701 PMCID: PMC10004236 DOI: 10.3390/jcm12051914] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/09/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
The pathophysiology, pathogenesis, histopathology, and immunopathology of medication-related osteonecrosis of the jaw (MRONJ) Stage 0 remain unclear, although 50% of MRONJ Stage 0 cases could progress to higher stages. The aim of this study was to investigate the effects of zoledronate (Zol) and anti-vascular endothelial cell growth factor A (VEGFA) neutralizing antibody (Vab) administration on polarization shifting of macrophage subsets in tooth extraction sockets by creating a murine model of MRONJ Stage 0-like lesions. Eight-week-old, female C57BL/6J mice were randomly divided into 4 groups: Zol, Vab, Zol/Vab combination, and vehicle control (VC). Subcutaneous Zol and intraperitoneal Vab administration were performed for 5 weeks with extraction of both maxillary first molars 3 weeks after drug administration. Euthanasia was conducted 2 weeks after tooth extraction. Maxillae, tibiae, femora, tongues, and sera were collected. Structural, histological, immunohistochemical, and biochemical analyses were comprehensively performed. Tooth extraction sites appeared to be completely healed in all groups. However, osseous healing and soft tissue healing of tooth extraction sites were quite different. The Zol/Vab combination significantly induced abnormal epithelial healing, and delayed connective tissue healing due to decreased rete ridge length and thickness of the stratum granulosum and due to decreased collagen production, respectively. Moreover, Zol/Vab significantly increased necrotic bone area with increased numbers of empty lacunae compared with Vab and VC. Most interestingly, Zol/Vab significantly increased the number of CD169+ osteal macrophages (osteomacs) in the bone marrow and decreased F4/80+ macrophages, with a slightly increased ratio of F4/80+CD38+ M1 macrophages compared to VC. These findings are the first to provide new evidence of the involvement of osteal macrophages in the immunopathology of MRONJ Stage 0-like lesions.
Collapse
|
15
|
Research Progress of Macrophages in Bone Regeneration. J Tissue Eng Regen Med 2023. [DOI: 10.1155/2023/1512966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Bone tissue regeneration plays an increasingly important role in contemporary clinical treatment. The reconstruction of bone defects remains a huge challenge for clinicians. Bone regeneration is regulated by the immune system, in which inflammation is an important regulating factor in bone formation and remodeling. As the main cells involved in inflammation, macrophages play a key role in osteogenesis by polarizing into different phenotypes during different stages of bone regeneration. Considering this, this review mainly summarizes the function of macrophage in bone regeneration based on mesenchymal stem cells (MSCs), osteoblasts, osteoclasts, and vascular cells. In conclusion, anti-inflammatory macrophages (M2) have a greater potentiality to promote bone regeneration than M0 and classically activated proinflammatory macrophages (M1). In the fracture and bone defect models, tissue engineering materials can induce the transition from M1 to M2, alter the bone microenvironment, and promote bone regeneration through interactions with bone-related cells and blood vessels. The review provides a further understanding of macrophage polarization behavior in the evolving field of bone immunology.
Collapse
|
16
|
Li D, Li X, Zhang J, Tang Z, Tian A. The immunomodulatory effect of IL-4 accelerates bone substitute material-mediated osteogenesis in aged rats via NLRP3 inflammasome inhibition. Front Immunol 2023; 14:1121549. [PMID: 37153554 PMCID: PMC10157059 DOI: 10.3389/fimmu.2023.1121549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Background Bone defect repair by implanting bone substitute materials has been a common clinical treatment. With the understanding of substance-immune system interactions and increasing evidence indicating that the post-implantation immune response determines the fate of bone substitute materials, active modulation of host macrophage polarization is considered a promising strategy. However, whether the same regulatory effects exist when an individual immune system is altered with aging is unclear. Methods In this study, we mechanistically investigated the effect of immunosenescence on the active regulation of macrophage polarization by establishing a cranial bone defect model in young and aged rats implanted with Bio-Oss®. Forty-eight young and 48 aged specific pathogen-free (SPF) male SD rats were randomly divided into two groups. In the experimental group, 20 μL of IL-4 (0.5 μg/mL) was injected locally on the third to seventh postoperative days, while an equal volume of PBS was injected in the control group. Specimens were collected at 1, 2, 6, and 12 weeks postoperatively, and bone regeneration at the defect site was evaluated by micro-CT, histomorphometry, immunohistochemistry, double-labeling immunofluorescence, and RT-qPCR. Results The application of exogenous IL-4 reduced activation of NLRP3 inflammasomes by promoting the polarization of M1 macrophages to M2 macrophages, thus promoting bone regeneration at the site of bone defects in aged rats. However, this effect was gradually weakened after the IL-4 intervention was discontinued. Conclusion Our data confirmed that a strategy to regulate macrophage polarization is also feasible under conditions of immunosenescence, i.e., the local inflammatory microenvironment can be regulated by reducing M1-type macrophages. However, further experiments are needed to determine an exogenous IL-4 intervention that can maintain a more sustained effect.
Collapse
Affiliation(s)
- Duchenhui Li
- Department of Prosthodontics and Implantology, School and Hospital of Stomatology of Guizhou Medical University, Guiyang, Guizhou, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology of Guizhou Medical University, Guiyang, China
- Department of Physiology and Pathology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Xiao Li
- Department of Oral and Maxillofacial Surgery, Guiyang Hospital of Stomatology, Guiyang, Guizhou, China
| | - Jie Zhang
- Department of Prosthodontics and Implantology, School and Hospital of Stomatology of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhenglong Tang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology of Guizhou Medical University, Guiyang, China
- Department of Physiology and Pathology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- *Correspondence: Ai Tian, ; Zhenglong Tang,
| | - Ai Tian
- Department of Prosthodontics and Implantology, School and Hospital of Stomatology of Guizhou Medical University, Guiyang, Guizhou, China
- *Correspondence: Ai Tian, ; Zhenglong Tang,
| |
Collapse
|
17
|
Bowles-Welch AC, Jimenez AC, Stevens HY, Frey Rubio DA, Kippner LE, Yeago C, Roy K. Mesenchymal stromal cells for bone trauma, defects, and disease: Considerations for manufacturing, clinical translation, and effective treatments. Bone Rep 2023. [DOI: 10.1016/j.bonr.2023.101656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
18
|
Arabiyat AS, Yeisley DJ, Güiza-Argüello VR, Qureshi F, Culibrk RA, Hahn J, Hahn MS. Effects of Stromal Cell Conditioned Medium and Antipurinergic Treatment on Macrophage Phenotype. Tissue Eng Part C Methods 2022; 28:656-671. [PMID: 36329666 PMCID: PMC9807257 DOI: 10.1089/ten.tec.2022.0123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
The immunomodulatory capacity of the human mesenchymal stromal cell (MSC) secretome has been a critical driver for the development of cell-free MSC products, such as conditioned medium (CM), for regenerative medicine applications. This is particularly true as cell-free MSC products present several advantages over direct autologous or allogeneic MSC delivery with respect to safety, manufacturability, and defined potency. Recently, significant effort has been placed into creating novel MSC CM formulations with an immunomodulatory capacity tailored for specific regenerative contexts. For instance, the immunoregulatory nature of MSC CM has previously been tuned through a number of cytokine-priming strategies. Herein, we propose an alternate method to tailor the immunomodulatory "phenotype" of cytokine-primed MSC CM through coupling with the pharmacological agent, suramin. Suramin interferes with the signaling of purines including extracellular adenosine triphosphate (ATP), which plays a critical role in the activation of the innate immune system after injury. Toward this end, human THP-1-derived macrophages were activated to a proinflammatory phenotype and treated with (1) unprimed/native MSC CM, (2) interferon-γ/tumor necrosis factor α-primed MSC CM (primed CM), (3) suramin alone, or (4) primed MSC CM and suramin (primed CM/suramin). Markers of key macrophage functions-cytokine secretion, autophagy, oxidative stress modulation, and activation/migration-were assessed. Consistent with previous literature, primed CM elevated macrophage secretion of several proinflammatory and pleiotropic cytokines relative to native CM; whereas addition of suramin imparted consistent shifts in terms of TNFα (↓), interleukin-10 (↓), and hepatocyte growth factor (↑) irrespective of CM. In addition, both primed CM and suramin, individually and combined, increased reactive oxygen species production relative to native CM, and addition of suramin to primed CM shifted levels of CX3CL1, a factor involved in ATP-associated macrophage regulation. Varimax rotation assessment of the secreted cytokine profiles confirmed that primed CM/suramin resulted in a THP-1 phenotypic shift away from the lipopolysaccharide-activated proinflammatory state that was distinct from that of primed CM or native CM alone. This altered primed CM/suramin-associated phenotype may prove beneficial for healing in certain regenerative contexts. These results may inform future work coupling antipurinergic treatments with MSC-derived therapies in regenerative medicine applications.
Collapse
Affiliation(s)
- Ahmad S. Arabiyat
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| | - Daniel J. Yeisley
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| | - Viviana R. Güiza-Argüello
- Department of Metallurgical Engineering and Materials Science, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Fatir Qureshi
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| | - Robert A. Culibrk
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| | - Juergen Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, New York, USA
| |
Collapse
|
19
|
Preconditioned Mesenchymal Stromal Cell-Derived Extracellular Vesicles (EVs) Counteract Inflammaging. Cells 2022; 11:cells11223695. [PMID: 36429124 PMCID: PMC9688039 DOI: 10.3390/cells11223695] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Inflammaging is one of the evolutionarily conserved mechanisms underlying aging and is defined as the long-term consequence of the chronic stimulation of the innate immune system. As macrophages are intimately involved in initiating and regulating the inflammatory process, their dysregulation plays major roles in inflammaging. The paracrine factors, and in particular extracellular vesicles (EVs), released by mesenchymal stromal cells (MSCs) retain immunoregulatory effects on innate and adaptive immune responses. In this paper, we demonstrate that EVs derived from MSCs preconditioned with hypoxia inflammatory cytokines exerted an anti-inflammatory role in the context of inflammaging. In this study, macrophages isolated from aged mice presented elevated pro-inflammatory factor levels already in basal conditions compared to the young counterpart, and this pre-activation status increased when cells were challenged with IFN-γ. EVs were able to attenuate the age-associated inflammation, inducing a decrease in the expression of TNF-α, iNOS, and the NADase CD38. Moreover, we demonstrate that EVs counteracted the mitochondrial dysfunction that affected the macrophages, reducing lipid peroxidation and hindering the age-associated impairment of mitochondrial complex I activity, oxygen consumption, and ATP synthesis. These results indicate that preconditioned MSC-derived EVs might be exploited as new anti-aging therapies in a variety of age-related diseases.
Collapse
|
20
|
Villalobos V, Garrido M, Reyes A, Fernández C, Diaz C, Torres VA, González PA, Cáceres M. Aging envisage imbalance of the periodontium: A keystone in oral disease and systemic health. Front Immunol 2022; 13:1044334. [PMID: 36341447 PMCID: PMC9630574 DOI: 10.3389/fimmu.2022.1044334] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/05/2022] [Indexed: 10/21/2023] Open
Abstract
Aging is a gradual and progressive deterioration of integrity across multiple organ systems that negatively affects gingival wound healing. The cellular responses associated with wound healing, such as collagen synthesis, cell migration, proliferation, and collagen contraction, have been shown to be lower in gingival fibroblasts (the most abundant cells from the connective gingival tissue) in aged donors than young donors. Cellular senescence is one of the hallmarks of aging, which is characterized by the acquisition of a senescence-associated secretory phenotype that is characterized by the release of pro-inflammatory cytokines, chemokines, growth factors, and proteases which have been implicated in the recruitment of immune cells such as neutrophils, T cells and monocytes. Moreover, during aging, macrophages show altered acquisition of functional phenotypes in response to the tissue microenvironment. Thus, inflammatory and resolution macrophage-mediated processes are impaired, impacting the progression of periodontal disease. Interestingly, salivary antimicrobial peptides, such as histatins, which are involved in various functions, such as antifungal, bactericidal, enamel-protecting, angiogenesis, and re-epithelization, have been shown to fluctuate with aging. Several studies have associated the presence of Porphyromonas gingivalis, a key pathogen related to periodontitis and apical periodontitis, with the progression of Alzheimer's disease, as well as gut, esophageal, and gastric cancers. Moreover, herpes simplex virus types 1 and 2 have been associated with the severity of periodontal disease, cardiovascular complications, and nervous system-related pathologies. This review encompasses the effects of aging on periodontal tissues, how P. gingivalis and HSV infections could favor periodontitis and their relationship with other pathologies.
Collapse
Affiliation(s)
- Verónica Villalobos
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Mauricio Garrido
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Antonia Reyes
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christian Fernández
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Catalina Diaz
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Vicente A. Torres
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica Cáceres
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Chile, Santiago, Chile
| |
Collapse
|
21
|
Lu YN, Wang L, Zhang YZ. The promising roles of macrophages in geriatric hip fracture. Front Cell Dev Biol 2022; 10:962990. [PMID: 36092716 PMCID: PMC9458961 DOI: 10.3389/fcell.2022.962990] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
As aging becomes a global burden, the incidence of hip fracture (HF), which is the most common fracture in the elderly population and can be fatal, is rapidly increasing, and its extremely high fatality rate places significant medical and financial burdens on patients. Fractures trigger a complex set of immune responses, and recent studies have shown that with aging, the immune system shows decreased activity or malfunctions in a process known as immune senescence, leading to disease and death. These phenomena are the reasons why elderly individuals typically exhibit chronically low levels of inflammation and increased rates of infection and chronic disease. Macrophages, which are key players in the inflammatory response, are critical in initiating the inflammatory response, clearing pathogens, controlling the innate and adaptive immune responses and repairing damaged tissues. Tissue-resident macrophages (TRMs) are widely present in tissues and perform immune sentinel and homeostatic functions. TRMs are combinations of macrophages with different functions and phenotypes that can be directly influenced by neighboring cells and the microenvironment. They form a critical component of the first line of defense in all tissues of the body. Immune system disorders caused by aging could affect the biology of macrophages and thus the cascaded immune response after fracture in various ways. In this review, we outline recent studies and discuss the potential link between monocytes and macrophages and their potential roles in HF in elderly individuals.
Collapse
Affiliation(s)
- Yi-ning Lu
- Department of Orthopedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ling Wang
- Department of Orthopedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Orthopedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Ying-ze Zhang, ; Ling Wang,
| | - Ying-ze Zhang
- Department of Orthopedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Ying-ze Zhang, ; Ling Wang,
| |
Collapse
|
22
|
Necula MG, Mazare A, Negrescu AM, Mitran V, Ozkan S, Trusca R, Park J, Schmuki P, Cimpean A. Macrophage-like Cells Are Responsive to Titania Nanotube Intertube Spacing-An In Vitro Study. Int J Mol Sci 2022; 23:3558. [PMID: 35408918 PMCID: PMC8998567 DOI: 10.3390/ijms23073558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/23/2022] Open
Abstract
With the introduction of a new interdisciplinary field, osteoimmunology, today, it is well acknowledged that biomaterial-induced inflammation is modulated by immune cells, primarily macrophages, and can be controlled by nanotopographical cues. Recent studies have investigated the effect of surface properties in modulating the immune reaction, and literature data indicate that various surface cues can dictate both the immune response and bone tissue repair. In this context, the purpose of the present study was to investigate the effects of titanium dioxide nanotube (TNT) interspacing on the response of the macrophage-like cell line RAW 264.7. The cells were maintained in contact with the surfaces of flat titanium (Ti) and anodic TNTs with an intertube spacing of 20 nm (TNT20) and 80 nm (TNT80), under standard or pro-inflammatory conditions. The results revealed that nanotube interspacing can influence macrophage response in terms of cell survival and proliferation, cellular morphology and polarization, cytokine/chemokine expression, and foreign body reaction. While the nanostructured topography did not tune the macrophages' differentiation into osteoclasts, this behavior was significantly reduced as compared to flat Ti surface. Overall, this study provides a new insight into how nanotubes' morphological features, particularly intertube spacing, could affect macrophage behavior.
Collapse
Affiliation(s)
- Madalina Georgiana Necula
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| | - Anca Mazare
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany; (A.M.); (S.O.); (P.S.)
- Advanced Institute for Materials Research (AIMR), National University Corporation Tohoku University (TU), Sendai 980-8577, Japan
| | - Andreea Mariana Negrescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| | - Valentina Mitran
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| | - Selda Ozkan
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany; (A.M.); (S.O.); (P.S.)
| | - Roxana Trusca
- Faculty of Engineering in Foreign Languages, University Politehnica of Bucharest, 313 Splaiul Indendentei, 060042 Bucharest, Romania;
| | - Jung Park
- Department of Pediatrics, Division of Molecular Pediatrics, University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Patrik Schmuki
- Department of Materials Science WW4-LKO, Friedrich-Alexander University, 91058 Erlangen, Germany; (A.M.); (S.O.); (P.S.)
- Regional Centre of Advanced Technologies and Materials, 78371 Olomouc, Czech Republic
- Department of Chemistry, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21569, Saudi Arabia
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania; (M.G.N.); (A.M.N.); (V.M.)
| |
Collapse
|
23
|
Hirata H, Zhang N, Ueno M, Barati D, Kushioka J, Shen H, Tsubosaka M, Toya M, Lin T, Huang E, Yao Z, Wu JY, Zwingenberger S, Yang F, Goodman SB. Ageing attenuates bone healing by mesenchymal stem cells in a microribbon hydrogel with a murine long bone critical-size defect model. Immun Ageing 2022; 19:14. [PMID: 35279175 PMCID: PMC8917642 DOI: 10.1186/s12979-022-00272-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/24/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Despite the high incidence of fractures and pseudoarthrosis in the aged population, a potential role for the use of mesenchymal stem cells (MSCs) in the treatment of bone defects in elderly patients has not been elucidated. Inflammation and the innate immune system, including macrophages, play crucial roles in the differentiation and activation of MSCs. We have developed lentivirus-transduced interleukin 4 (IL4) over-expressing MSCs (IL4-MSCs) to polarize macrophages to an M2 phenotype to promote bone healing in an established young murine critical size bone defect model. In the current study, we explore the potential of IL4-MSCs in aged mice. METHODS A 2 mm femoral diaphyseal bone defect was created and fixed with an external fixation device in 15- to 17-month-old male and female BALB/c mice. Microribbon (µRB) scaffolds (Sc) with or without encapsulation of MSCs were implanted in the defect sites. Accordingly, the mice were divided into three treatment groups: Sc-only, Sc + MSCs, and Sc + IL4-MSCs. Mice were euthanized six weeks after the surgery; subsequently, MicroCT (µCT), histochemical and immunohistochemical analyses were performed. RESULTS µCT analysis revealed that bone formation was markedly enhanced in the IL4-MSC group. Compared with the Sc-only, the amount of new bone increased in the Sc + MSCs and Sc + IL4-MSC groups. However, no bridging of bone was observed in all groups. H&E staining showed fibrous tissue within the defect in all groups. Alkaline phosphatase (ALP) staining was increased in the Sc + IL4-MSC group. The Sc + IL4-MSCs group showed a decrease in the number of M1 macrophages and an increase in the number of M2 macrophages, with a significant increase in the M2/M1 ratio. DISCUSSION IL4 promotes macrophage polarization to an M2 phenotype, facilitating osteogenesis and vasculogenesis. The addition of IL4-MSCs in the µRB scaffold polarized macrophages to an M2 phenotype and increased bone formation; however, complete bone bridging was not observed in any specimens. These results suggest that IL4-MSCs are insufficient to heal a critical size bone defect in aged mice, as opposed to younger animals. Additional therapeutic strategies are needed in this challenging clinical scenario.
Collapse
Affiliation(s)
- Hirohito Hirata
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA.,Department of Orthopaedic Surgery, Saga University, Saga, Japan
| | - Danial Barati
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Junichi Kushioka
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Ejun Huang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Joy Y Wu
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Stefan Zwingenberger
- University Center for Orthopaedics, Traumatology, and Plastic Surgery, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA.,Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA. .,Department of Bioengineering, Stanford University, Stanford, California, USA.
| |
Collapse
|
24
|
Afroz SF, Condon ND, Sweet MJ, Kapetanovic R. Quantifying Regulated Mitochondrial Fission in Macrophages. Methods Mol Biol 2022; 2523:281-301. [PMID: 35759204 DOI: 10.1007/978-1-0716-2449-4_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Mitochondria have co-evolved with eukaryotic cells for more than a billion years, becoming an important cog in their machinery. They are best known for being tasked with energy generation through the production of adenosine triphosphate, but they also have roles in several other cellular processes, for example, immune and inflammatory responses. Mitochondria have important functions in macrophages, key innate immune cells that detect pathogens and drive inflammation. Mitochondrial activity is influenced by the highly dynamic nature of the mitochondrial network, which alternates between interconnected tubular and fragmented forms. The dynamic balance between this interconnected fused network and fission-mediated mitochondrial fragmentation modulates inflammatory responses such as production of cytokines and mitochondrial reactive oxygen species. Here we describe methods to differentiate mouse bone marrow cells into macrophages and the use of light microscopy, electron microscopy, flow cytometry, and Western blotting to quantify regulated mitochondrial dynamics in these differentiated macrophages.
Collapse
Affiliation(s)
- Syeda Farhana Afroz
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Nicholas D Condon
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia.
- IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, QLD, Australia.
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.
| | - Ronan Kapetanovic
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia.
- IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, QLD, Australia.
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.
| |
Collapse
|
25
|
Abstract
Two vasculitides, giant cell arteritis (GCA) and Takayasu arteritis (TAK), are recognized as autoimmune and autoinflammatory diseases that manifest exclusively within the aorta and its large branches. In both entities, the age of the affected host is a critical risk factor. TAK manifests during the 2nd-4th decade of life, occurring while the immune system is at its height of performance. GCA is a disease of older individuals, with infrequent cases during the 6th decade and peak incidence during the 8th decade of life. In both vasculitides, macrophages and T cells infiltrate into the adventitia and media of affected vessels, induce granulomatous inflammation, cause vessel wall destruction, and reprogram vascular cells to drive adventitial and neointimal expansion. In GCA, abnormal immunity originates in an aged immune system and evolves within the aged vascular microenvironment. One hallmark of the aging immune system is the preferential loss of CD8+ T cell function. Accordingly, in GCA but not in TAK, CD8+ effector T cells play a negligible role and anti-inflammatory CD8+ T regulatory cells are selectively impaired. Here, we review current evidence of how the process of immunosenescence impacts the risk for GCA and how fundamental differences in the age of the immune system translate into differences in the granulomatous immunopathology of TAK versus GCA.
Collapse
|
26
|
Sun X, Gao J, Meng X, Lu X, Zhang L, Chen R. Polarized Macrophages in Periodontitis: Characteristics, Function, and Molecular Signaling. Front Immunol 2021; 12:763334. [PMID: 34950140 PMCID: PMC8688840 DOI: 10.3389/fimmu.2021.763334] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/04/2021] [Indexed: 12/23/2022] Open
Abstract
Periodontitis (PD) is a common chronic infectious disease. The local inflammatory response in the host may cause the destruction of supporting periodontal tissue. Macrophages play a variety of roles in PD, including regulatory and phagocytosis. Moreover, under the induction of different factors, macrophages polarize and form different functional phenotypes. Among them, M1-type macrophages with proinflammatory functions and M2-type macrophages with anti-inflammatory functions are the most representative, and both of them can regulate the tendency of the immune system to exert proinflammatory or anti-inflammatory functions. M1 and M2 macrophages are involved in the destructive and reparative stages of PD. Due to the complex microenvironment of PD, the dynamic development of PD, and various local mediators, increasing attention has been given to the study of macrophage polarization in PD. This review summarizes the role of macrophage polarization in the development of PD and its research progress.
Collapse
Affiliation(s)
- Xiaoyu Sun
- *Correspondence: Lei Zhang, ; Xiaoyu Sun,
| | | | | | | | - Lei Zhang
- Key Laboratory of Oral Diseases Research of Anhui Province, Department of Periodontology, Stomatologic Hospital & College, Anhui Medical University, Hefei, China
| | | |
Collapse
|
27
|
Schlundt C, Fischer H, Bucher CH, Rendenbach C, Duda GN, Schmidt-Bleek K. The multifaceted roles of macrophages in bone regeneration: A story of polarization, activation and time. Acta Biomater 2021; 133:46-57. [PMID: 33974949 DOI: 10.1016/j.actbio.2021.04.052] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/26/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
To present knowledge, macrophages are found in all tissues of the human body. They are a cell population with high plasticity which come with a multitude of functions which appear to be adapted to the respective tissue niche and micro-environment in which they reside. Bone harbors multiple macrophage subpopulations, with the osteoclasts as classical representative of a bone resorbing cells and osteomacs as a bone tissue resident macrophage first described by the expression of F4/80. Both subtypes are found throughout all phases in bone healing. In vivo data on bone regeneration have demonstrated their essential role in initiating the healing cascade (inflammatory phase) but also of the later phases of healing (e.g. endochondral and intramembranous bone formation). To participate in such diverse processes macrophages have to be highly plastic in their functionality. Thus, the widely used M1/M2 paradigm to distinguish macrophage subpopulations may not mirror the comprehensive role of the dynamics of macrophage plasticity. From a clinical perspective it is especially relevant to distinguish what drives macrophages in impaired healing scenarios, implant loosening or infections, where their specific role of a misbalanced inflammatory setting is so far only partially known. With this review we aim at illustrating current knowledge and gaps of knowledge on macrophage plasticity and function during the cascades of regeneration and reconstitution of bone tissue. We propose aspects of the known biological mechanisms of macrophages and their specific subsets that might serve as targets to control their function in impaired healing and eventually support a scar-free regeneration. STATEMENT OF SIGNIFICANCE: Macrophages are essential for successful regeneration. In scar-free healing such as in bone, a complete failure of healing was shown if macrophages were depleted; the M1/M2 switch appears to be key to the progression from pro-inflammation to regeneration. However, experimental data illustrate that the classical M1/M2 paradigm does not completely mirror the complexity of observed macrophage functions during bone healing and thus demands a broader perspective. Within this review we discuss the high degree of plasticity of macrophages and the relevant contribution of the different and more specific M2 subtypes (M2a-M2f) during (bone) regeneration. It summarizes the versatile roles of macrophages in skeletal regeneration and thereby highlights potential target points for immunomodulatory approaches to enable or even foster bone repair.
Collapse
|
28
|
Yang Y, Zhang H, Komasa S, Morimoto Y, Sekino T, Kawazoe T, Okazaki J. UV/ozone irradiation manipulates immune response for antibacterial activity and bone regeneration on titanium. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112377. [PMID: 34579896 DOI: 10.1016/j.msec.2021.112377] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
The immunomodulatory antibacterial activity and osteoimmunomodulatory properties of implantable biomaterials significantly influence bone regeneration. Various types of ultraviolet (UV) instrument are currently in use to greatly enhance the antibacterial activity and osteoconductive capability of titanium, it remains unclear how UV treatment modulates immune response. Compared to traditional UV treatment, the combination of low-dose ozone with UV irradiation is considered a new option to give benefits to surface modification and reduce the drawbacks of UV and ozone individually. Herein, the aim of this study was to elucidate the immune-modulatory properties of macrophages on UV/ozone-irradiated titanium that serve as defense against S. aureus and the crosstalk between immune cells and osteoblasts. Three different cell and bacteria co-culture systems were developed in order to investigate the race between host cells and bacteria to occupy the surface. In vitro immunological experiments indicated that UV/ozone irradiation significantly enhanced the phagocytic and bactericidal activity of macrophages against S. aureus. Further, in vitro and in vivo studies evidenced the favorable osteoimmune environment for osteogenic differentiation and bone formation. This research suggests vital therapeutic potential of UV/ozone irradiation for preventing the biomaterial-associated infections and achieving favorable bone formation simultaneously.
Collapse
Affiliation(s)
- Yuanyuan Yang
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1 Kuzuha-hanazono-cho, Hirakata, Osaka 573-1121, Japan.
| | - Honghao Zhang
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1 Kuzuha-hanazono-cho, Hirakata, Osaka 573-1121, Japan.
| | - Satoshi Komasa
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1 Kuzuha-hanazono-cho, Hirakata, Osaka 573-1121, Japan
| | - Yukihiro Morimoto
- The Institute of Scientific and Industrial Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Tohru Sekino
- The Institute of Scientific and Industrial Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takayoshi Kawazoe
- Osaka Dental University, 8-1 Kuzuha-hanazono-cho, Hirakata, Osaka 573-1121, Japan
| | - Joji Okazaki
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1 Kuzuha-hanazono-cho, Hirakata, Osaka 573-1121, Japan
| |
Collapse
|
29
|
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: 70] [Impact Index Per Article: 23.3] [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
|
30
|
Arvind V, Huang AH. Reparative and Maladaptive Inflammation in Tendon Healing. Front Bioeng Biotechnol 2021; 9:719047. [PMID: 34350166 PMCID: PMC8327090 DOI: 10.3389/fbioe.2021.719047] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/28/2021] [Indexed: 12/26/2022] Open
Abstract
Tendon injuries are common and debilitating, with non-regenerative healing often resulting in chronic disease. While there has been considerable progress in identifying the cellular and molecular regulators of tendon healing, the role of inflammation in tendon healing is less well understood. While inflammation underlies chronic tendinopathy, it also aids debris clearance and signals tissue repair. Here, we highlight recent findings in this area, focusing on the cells and cytokines involved in reparative inflammation. We also discuss findings from other model systems when research in tendon is minimal, and explore recent studies in the treatment of human tendinopathy to glean further insights into the immunobiology of tendon healing.
Collapse
Affiliation(s)
- Varun Arvind
- Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alice H. Huang
- Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Orthopedic Surgery, Columbia University, New York, NY, United States
| |
Collapse
|
31
|
Kang K, Xia A, Meng F, Chunyu J, Sun X, Ren G, Yu D, Jiang X, Tang L, Xiao W, Li D. FGF21 alleviates chronic inflammatory injury in the aging process through modulating polarization of macrophages. Int Immunopharmacol 2021; 96:107634. [PMID: 33872851 DOI: 10.1016/j.intimp.2021.107634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 01/14/2023]
Abstract
Previous studies reported that FGF21 prolongs life span and delays the body senescence, but the mechanism is not clear. The present study was designed to investigate the effects of FGF21 on hepatic senescence in aging mice and further research the mechanism. The 14-month-old male mice were administered with PBS, FGF21 or metformin once daily for 6 months. Results showed that FGF21 alleviated liver injury and inhibited accumulation of senescence markers SASP, P53 and P16 in the livers of aging mice. Subsequently we found that the aging mice treated by FGF21 showed transition of type 1 macrophages (M1) to type 2 macrophages (M2) in the livers. Next, we used THP-1 macrophages triggered by LPS to study effects of FGF21 on macrophages. Macrophages triggered by LPS exhibited features of M1, but the addition of FGF21 decreased the expression of M1 markers, and promoted the macrophages to exhibit features of M2. Results showed that the effects of FGF21 on macrophages were associated with the AMPK pathway. After adding AMPK inhibitor, the effects of FGF21 were inhibited, which was associated with the NF-κB signaling pathway. Finally, co-culturing differentiated macrophages and hepatocytes, we found that the large amount of pro-inflammatory factors such as IL-6 promoted hepatocyte senescence, which exhibited enhanced P53, P16 and β-galactosidase. This was contrary to hepatocytes co-cultured with macrophages treated by FGF21. These results indicate that FGF21 alleviates hepatic senescence injury by modulating the polarization of macrophages through the AMPK /NF-κB signaling pathway.
Collapse
Affiliation(s)
- Kai Kang
- Northeast Agricultural University, Harbin, China.
| | - Anran Xia
- Northeast Agricultural University, Harbin, China.
| | - Fanrui Meng
- Northeast Agricultural University, Harbin, China.
| | - Jian Chunyu
- Northeast Agricultural University, Harbin, China.
| | - Xu Sun
- Northeast Agricultural University, Harbin, China.
| | - Guiping Ren
- Northeast Agricultural University, Harbin, China.
| | - Dan Yu
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical CO. LTD, Lianyungang, Jiangsu, China.
| | | | - Lei Tang
- Harbin Weike Biotechnology CO. LTD, Harbin, China.
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical CO. LTD, Lianyungang, Jiangsu, China.
| | - Deshan Li
- Northeast Agricultural University, Harbin, China.
| |
Collapse
|
32
|
Treatment with a long-acting chimeric CSF1 molecule enhances fracture healing of healthy and osteoporotic bones. Biomaterials 2021; 275:120936. [PMID: 34303178 DOI: 10.1016/j.biomaterials.2021.120936] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/28/2021] [Accepted: 05/25/2021] [Indexed: 12/27/2022]
Abstract
Macrophage-targeted therapies, including macrophage colony-stimulating factor 1 (CSF1), have been shown to have pro-repair impacts post-fracture. Preclinical/clinical applications of CSF1 have been expedited by development of chimeric CSF1-Fc which has extended circulating half-life. Here, we used mouse models to investigate the bone regenerative potential of CSF1-Fc in healthy and osteoporotic fracture. We also explored whether combination of CSF1-Fc with interleukin (IL)-4 provided additional fracture healing benefit in osteopenic bone. Micro-computed tomography, in situ histomorphometry, and bone mechanical parameters were used to assess systemic impacts of CSF1-Fc therapy in naive mice (male and female young, adult and geriatric). An intermittent CSF1-Fc regimen was optimized to mitigate undesirable impacts on bone resorption and hepatosplenomegaly, irrespective of age or gender. The intermittent CSF1-Fc regimen was tested in a mid-diaphyseal femoral fracture model in healthy bones with treatment initiated 1-day post-fracture. Weekly CSF1-Fc did not impact osteoclasts but increased osteal macrophages and improved fracture strength. Importantly, this treatment regimen also improved fracture union and strength in an ovariectomy-model of delayed fracture repair. Combining CSF1-Fc with IL-4 initiated 1-week post-fracture reduced the efficacy of CSF1-Fc. This study describes a novel strategy to specifically achieve bone regenerative actions of CSF1-Fc that has the potential to alleviate fragility fracture morbidity and mortality.
Collapse
|
33
|
Hagen KM, Ousman SS. Aging and the immune response in diabetic peripheral neuropathy. J Neuroimmunol 2021; 355:577574. [PMID: 33894676 DOI: 10.1016/j.jneuroim.2021.577574] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/09/2021] [Accepted: 04/10/2021] [Indexed: 12/27/2022]
Abstract
A large proportion of older individuals with diabetes go on to develop diabetic peripheral neuropathy (DPN). DPN is associated with an increase in inflammatory cells within the peripheral nerve, activation of nuclear factor kappa-light-chain-enhancer of activated B cells and receptors for advanced glycation end products/advanced glycation end products pathways, aberrant cytokine expression, oxidative stress, ischemia, as well as pro-inflammatory changes in the bone marrow; all processes that may be exacerbated with age. We review the immunological features of DPN and discuss whether age-related changes in relevant immunological areas may contribute to age being a risk factor for DPN.
Collapse
Affiliation(s)
- Kathleen M Hagen
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Shalina S Ousman
- Departments of Clinical Neurosciences and Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
34
|
Lu RJ, Wang EK, Benayoun BA. Functional genomics of inflamm-aging and immunosenescence. Brief Funct Genomics 2021; 21:43-55. [PMID: 33690792 DOI: 10.1093/bfgp/elab009] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
The aging population is at a higher risk for age-related diseases and infections. This observation could be due to immunosenescence: the decline in immune efficacy of both the innate and the adaptive immune systems. Age-related immune decline also links to the concept of 'inflamm-aging,' whereby aging is accompanied by sterile chronic inflammation. Along with a decline in immune function, aging is accompanied by a widespread of 'omics' remodeling. Transcriptional landscape changes linked to key pathways of immune function have been identified across studies, such as macrophages having decreased expression of genes associated to phagocytosis, a major function of macrophages. Therefore, a key mechanism underlying innate immune cell dysfunction during aging may stem from dysregulation of youthful genomic networks. In this review, we discuss both molecular and cellular phenotypes of innate immune cells that contribute to age-related inflammation.
Collapse
Affiliation(s)
- Ryan J Lu
- Leonard Davis School of Gerontology at the University of Southern California
| | - Emily K Wang
- Leonard Davis School of Gerontology at the University of Southern California
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology at the University of Southern California
| |
Collapse
|
35
|
McGowan LM, Kague E, Vorster A, Newham E, Cross S, Hammond CL. Wnt16 Elicits a Protective Effect Against Fractures and Supports Bone Repair in Zebrafish. JBMR Plus 2021; 5:e10461. [PMID: 33778326 PMCID: PMC7990157 DOI: 10.1002/jbm4.10461] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/09/2020] [Accepted: 01/03/2021] [Indexed: 12/23/2022] Open
Abstract
Bone homeostasis is a dynamic, multicellular process that is required throughout life to maintain bone integrity, prevent fracture, and respond to skeletal damage. WNT16 has been linked to bone fragility and osteoporosis in human genome wide‐association studies, as well as the functional hematopoiesis of leukocytes in vivo. However, the mechanisms by which WNT16 promotes bone health and repair are not fully understood. In this study, CRISPR‐Cas9 was used to generate mutant zebrafish lacking Wnt16 (wnt16−/−) to study its effect on bone dynamically. The wnt16 mutants displayed variable tissue mineral density (TMD) and were susceptible to spontaneous fractures and the accumulation of bone calluses at an early age. Fractures were induced in the lepidotrichia of the caudal fins of wnt16−/− and WT zebrafish; this model was used to probe the mechanisms by which Wnt16 regulates skeletal and immune cell dynamics in vivo. In WT fins, wnt16 expression increased significantly during the early stages for bone repair. Mineralization of bone during fracture repair was significantly delayed in wnt16 mutants compared with WT zebrafish. Surprisingly, there was no evidence that the recruitment of innate immune cells to fractures or soft callus formation was altered in wnt16 mutants. However, osteoblast recruitment was significantly delayed in wnt16 mutants postfracture, coinciding with precocious activation of the canonical Wnt signaling pathway. In situ hybridization suggests that canonical Wnt‐responsive cells within fractures are osteoblast progenitors, and that osteoblast differentiation during bone repair is coordinated by the dynamic expression of runx2a and wnt16. This study highlights zebrafish as an emerging model for functionally validating osteoporosis–associated genes and investigating fracture repair dynamically in vivo. Using this model, it was found that Wnt16 protects against fracture and supports bone repair, likely by modulating canonical Wnt activity via runx2a to facilitate osteoblast differentiation and bone matrix deposition. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Lucy M McGowan
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Erika Kague
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Alistair Vorster
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Elis Newham
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Stephen Cross
- Wolfson Bioimaging Facility University of Bristol Bristol UK
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| |
Collapse
|
36
|
Chen M, Zhang Y, Zhou P, Liu X, Zhao H, Zhou X, Gu Q, Li B, Zhu X, Shi Q. Substrate stiffness modulates bone marrow-derived macrophage polarization through NF-κB signaling pathway. Bioact Mater 2020; 5:880-890. [PMID: 32637751 PMCID: PMC7332470 DOI: 10.1016/j.bioactmat.2020.05.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/06/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023] Open
Abstract
The stiffness of the extracellular matrix (ECM) plays an important role in regulating the cellular programming. However, the mechanical characteristics of ECM affecting cell differentiation are still under investigated. Herein, we aimed to study the effect of ECM substrate stiffness on macrophage polarization. We prepared polyacrylamide hydrogels with different substrate stiffness, respectively. After the hydrogels were confirmed to have a good biocompatibility, the bone marrow-derived macrophages (BMMs) from mice were incubated on the hydrogels. With simulated by the low substrate stiffness, BMMs displayed an enhanced expression of CD86 on the cell surface and production of reactive oxygen species (ROS) in cells, and secreted more IL-1β and TNF-α in the supernatant. On the contrary, stressed by the medium stiffness, BMMs expressed more CD206, produced less ROS, and secreted more IL-4 and TGF-β. In vivo study by delivered the hydrogels subcutaneously in mice, more CD68+CD86+ cells around the hydrogels with the low substrate stiffness were observed while more CD68+CD206+ cells near by the middle stiffness hydrogels. In addition, the expressions of NIK, phosphorylated p65 (pi-p65) and phosphorylated IκB (pi-IκB) were significantly increased after stimulation with low stiffness in BMMs. Taken together, these findings demonstrated that substrate stiffness could affect macrophages polarization. Low substrate stiffness promoted BMMs to shift to classically activated macrophages (M1) and the middle one to alternatively activated macrophages (M2), through modulating ROS-initiated NF-κB pathway. Therefore, we anticipated ECM-based substrate stiffness with immune modulation would be under consideration in the clinical applications if necessary.
Collapse
Affiliation(s)
- Mimi Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Yu Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Pinghui Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Xingzhi Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Huan Zhao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Xichao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Qiaoli Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Bin Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Xuesong Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Qin Shi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, 199 Renai Road, Suzhou, 215123, PR China
| |
Collapse
|
37
|
Wasityastuti W, Habib NA, Sari DCR, Arfian N. Effects of low and moderate treadmill exercise on liver of d-galactose-exposed aging rat model. Physiol Rep 2020; 7:e14279. [PMID: 31724278 PMCID: PMC6854106 DOI: 10.14814/phy2.14279] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
Aging increases liver susceptibility to diseases and it causes inflammation in liver tissue which can lead to fibrosis. Studies suggest that aging is caused by the accumulation of free radicals. Lack of physical activity can lower hormone levels and increase free radicals that can accelerate the aging process. Hence, physical activity is very important to maintain functions of organs. This research was aimed to study the effects of low and moderate treadmill exercise on d‐Galactose‐exposed aging rat model by evaluating the degree of hepatic fibrosis, number of M1 and M2, and M1/M2 ratio. Twenty‐four 3‐month‐old male Wistar aging model rats were randomly divided into four groups, that is, three treatment groups with daily 300 mg kgBW−1d‐Galactose injection administrated intraperitoneally for 4 weeks and 1 control group with normal saline injection. Two of the d‐Galactose treated groups were given low and moderate treadmill exercise for 4 weeks. It was concluded that low intensity treadmill exercise significantly lowered the degree of d‐Galactose‐exposed hepatic fibrosis, and moderate treadmill exercise was able to restore the injured liver tissue back to the non‐aging state. Administration of d‐Galactose causes inflammation marked by the elevated number of M1 and M2 macrophages. Moderate treadmill exercise drove M1/M2 ratio back to the control condition.
Collapse
Affiliation(s)
- Widya Wasityastuti
- Department of Physiology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Nurfatma A Habib
- Master in Biomedical Sciences, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Dwi C R Sari
- Department of Anatomy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Nur Arfian
- Department of Anatomy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
38
|
Huang R, Vi L, Zong X, Baht GS. Maresin 1 resolves aged-associated macrophage inflammation to improve bone regeneration. FASEB J 2020; 34:13521-13532. [PMID: 32794246 PMCID: PMC7719599 DOI: 10.1096/fj.202001145r] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022]
Abstract
Inflammaging is associated with poor tissue regeneration observed in advanced age. Specifically, protracted inflammation after acute injury has been associated with decreased bone fracture healing and increased rates of nonunion in elderly patients. Here, we investigated the efficacy of using Maresin 1 (MaR1), an omega-3 fatty acid-derived pro-resolving agent, to resolve inflammation after tibial fracture injury and subsequently improving aged bone healing. Aged (24-month-old mice) underwent tibial fracture surgery and were either treated with vehicle or MaR1 3 days after injury. Fracture calluses were harvested 7 days, 14 days, 21 days, and 28 days after injury to investigate inflammatory response, cartilage development, bone deposition, and mechanical integrity, respectively. Healing bones from MaR1-treated mice displayed decreased cartilage formation and increased bone deposition which resulted in increased structural stiffness and increased force to fracture in the later stages of repair. In the early stages, MaR1 treatment decreased the number of pro-inflammatory macrophages within the fracture callus and decreased the level of inflammatory biomarkers in circulation. In tissue culture models, MaR1 treatment of bone marrow-derived macrophages from aged mice protected cells form a pro-inflammatory phenotype and induced an anti-inflammatory fate. Furthermore, the secretome of MaR1-treated bone marrow-derived macrophages was identified as osteoinductive, enhancing osteoblast differentiation of bone marrow stromal cells. Our findings here identify resolution of inflammation, and MaR1 itself, to be a point of intervention to improve aged bone healing.
Collapse
Affiliation(s)
- Rong Huang
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA.,Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Linda Vi
- Division of Physical Medicine and Rehabilitation, University of Toronto, Toronto, Canada
| | - Xiaohua Zong
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA.,Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Gurpreet S Baht
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA.,Department of Orthopaedic Surgery, Duke University, Durham, NC, USA.,Department of Pathology, Duke University, Durham, NC, USA
| |
Collapse
|
39
|
Macrophage Immunometabolism and Inflammaging: Roles of Mitochondrial Dysfunction, Cellular Senescence, CD38, and NAD. ACTA ACUST UNITED AC 2020; 2:e200026. [PMID: 32774895 PMCID: PMC7409778 DOI: 10.20900/immunometab20200026] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aging is a complex process that involves dysfunction on multiple levels, all of which seem to converge on inflammation. Macrophages are intimately involved in initiating and resolving inflammation, and their dysregulation with age is a primary contributor to inflammaging—a state of chronic, low-grade inflammation that develops during aging. Among the age-related changes that occur to macrophages are a heightened state of basal inflammation and diminished or hyperactive inflammatory responses, which seem to be driven by metabolic-dependent epigenetic changes. In this review article we provide a brief overview of mitochondrial functions and age-related changes that occur to macrophages, with an emphasis on how the inflammaging environment, senescence, and NAD decline can affect their metabolism, promote dysregulation, and contribute to inflammaging and age-related pathologies.
Collapse
|
40
|
Tang Y, Tong X, Conrad B, Yang F. Injectable and in situ crosslinkable gelatin microribbon hydrogels for stem cell delivery and bone regeneration in vivo. Theranostics 2020; 10:6035-6047. [PMID: 32483436 PMCID: PMC7254995 DOI: 10.7150/thno.41096] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 04/27/2020] [Indexed: 12/20/2022] Open
Abstract
Rationale: Injectable matrices are highly desirable for stem cell delivery. Previous research has highlighted the benefit of scaffold macroporosity in enhancing stem cell survival and bone regeneration in vivo. However, there remains a lack of injectable and in situ crosslinkable macroporous matrices for stem cell delivery to achieve fast bone regeneration in immunocompetent animal model. The goal of this study is to develop an injectable gelatin-based μRB hydrogel supporting direct cell encapsulation that is available in clinics as macroporous matrices to enhance adipose-derived stromal cell (ASC) survival, engraftment and accelerate bone formation in craniofacial defect mouse. Methods: Injectable and in situ crosslinkable gelatin microribbon (μRB)-based macroporous hydrogels were developed by wet-spinning. Injectability was optimized by varying concentration of glutaraldehyde for intracrosslinking of μRB shape, and fibrinogen coating. The efficacy of injectable μRBs to support ASCs delivery and bone regeneration were further assessed in vivo using an immunocompetent mouse cranial defect model. ASCs survival was evaluated by bioluminescent imaging and bone regeneration was assessed by micro-CT. The degradation and biocompatibility were determined by histological analysis. Results: We first optimized injectability by varying concentration of glutaraldehyde used to fix gelatin μRBs. The injectable μRB formulation were subsequently coated with fibrinogen, which allows in situ crosslinking by thrombin. Fluorescence imaging and histology showed majority of μRBs degraded by the end of 3 weeks. Injectable μRBs supported comparable level of ASC proliferation and bone regeneration as implantable prefabricated μRB controls. Adding low dosage of BMP2 (100 ng per scaffold) with ASCs substantially accelerated the speed of mineralized bone regeneration, with 90% of the bone defect refilled by week 8. Immunostaining showed M1 (pro-inflammatory) macrophages were recruited to the defect at day 3, and was replaced by M2 (anti-inflammatory) macrophages by week 2. Adding μRBs or BMP2 did not alter macrophage response. Injectable µRBs supported vascularization, and BMP-2 further enhanced vascularization. Conclusions: Our results demonstrated that µRB-based scaffolds enhanced ASC survival and accelerated bone regeneration after injection into critical sized cranial defect mouse. Such injectable µRB-based scaffold can provide a versatile biomaterial for delivering various stem cell types and enhancing tissue regeneration.
Collapse
|
41
|
Clark D, Brazina S, Yang F, Hu D, Hsieh CL, Niemi EC, Miclau T, Nakamura MC, Marcucio R. Age-related changes to macrophages are detrimental to fracture healing in mice. Aging Cell 2020; 19:e13112. [PMID: 32096907 PMCID: PMC7059136 DOI: 10.1111/acel.13112] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/05/2019] [Accepted: 01/17/2020] [Indexed: 12/12/2022] Open
Abstract
The elderly population suffers from higher rates of complications during fracture healing that result in increased morbidity and mortality. Inflammatory dysregulation is associated with increased age and is a contributing factor to the myriad of age-related diseases. Therefore, we investigated age-related changes to an important cellular regulator of inflammation, the macrophage, and the impact on fracture healing outcomes. We demonstrated that old mice (24 months) have delayed fracture healing with significantly less bone and more cartilage compared to young mice (3 months). The quantity of infiltrating macrophages into the fracture callus was similar in old and young mice. However, RNA-seq analysis demonstrated distinct differences in the transcriptomes of macrophages derived from the fracture callus of old and young mice, with an up-regulation of M1/pro-inflammatory genes in macrophages from old mice as well as dysregulation of other immune-related genes. Preventing infiltration of the fracture site by macrophages in old mice improved healing outcomes, with significantly more bone in the calluses of treated mice compared to age-matched controls. After preventing infiltration by macrophages, the macrophages remaining within the fracture callus were collected and examined via RNA-seq analysis, and their transcriptome resembled macrophages from young calluses. Taken together, infiltrating macrophages from old mice demonstrate detrimental age-related changes, and depleting infiltrating macrophages can improve fracture healing in old mice.
Collapse
Affiliation(s)
- Daniel Clark
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
- Division of PeriodontologyDepartment of Orofacial SciencesSchool of DentistryUniversity of California San FranciscoSan FranciscoCAUSA
| | - Sloane Brazina
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
| | - Frank Yang
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
| | - Diane Hu
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
| | - Christine L. Hsieh
- Division of RheumatologyDepartment of MedicineSan Francisco VA Health Care SystemSan FranciscoCAUSA
| | - Erene C. Niemi
- Division of RheumatologyDepartment of MedicineSan Francisco VA Health Care SystemSan FranciscoCAUSA
| | - Theodore Miclau
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
| | - Mary C. Nakamura
- Division of RheumatologyDepartment of MedicineSan Francisco VA Health Care SystemSan FranciscoCAUSA
| | - Ralph Marcucio
- Department of Orthopaedic SurgerySchool of MedicineOrthopaedic Trauma InstituteZuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCAUSA
| |
Collapse
|
42
|
Nasirzade J, Kargarpour Z, Hasannia S, Strauss FJ, Gruber R. Platelet-rich fibrin elicits an anti-inflammatory response in macrophages in vitro. J Periodontol 2020; 91:244-252. [PMID: 31376159 PMCID: PMC7065136 DOI: 10.1002/jper.19-0216] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/31/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Platelet-rich fibrin (PRF) serves as a reservoir of bioactive molecules to support wound healing and bone regeneration. The beneficial action of PRF might involve macrophage polarization from proinflammatory M1 toward pro-resolving M2 phenotypes. This study aims to evaluate the effect of PRF on macrophage polarization. METHODS Murine primary macrophages and RAW 264.7 cells were exposed to saliva and lipopolysaccharides (LPS) with and without PRF lysates obtained by repeated freeze-thawing or the secretome of PRF membranes, termed PRF conditioned medium. The expression of the M1 marker genes interleukin 1β (IL1β) and interleukin 6 (IL6) along with the M2 markers arginase-1 and chitinase-like 3 (Chil3 or YM1) were evaluated by real time polymerase chain reaction. Immunoassay and immunofluorescence staining were performed for IL6 and p65 translocation, a subunit nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), respectively. RESULTS We report here that PRF lysates and PRF conditioned medium, the latter containing the secretome, greatly decreased the proinflammatory response of primary macrophages and RAW 264.7 cells as indicated by the expression of IL1β and IL6. The anti-inflammatory activity of PRF lysates was further confirmed by IL6 immunoassay. Moreover, PRF lysates suppressed the translocation of p65 from the cytoplasm into the nucleus after incubation with saliva. In support of M2 polarization, PRF lysates and PRF conditioned medium enhanced the expression of arginase-1 and YM1 in primary macrophages. CONCLUSION Our results indicate that PRF holds an anti-inflammatory activity and shifts the macrophage polarization from an M1 toward an M2 phenotype.
Collapse
Affiliation(s)
- Jila Nasirzade
- Department of Oral BiologyMedical University of ViennaViennaAustria
- Department of BiochemistryFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Zahra Kargarpour
- Department of Oral BiologyMedical University of ViennaViennaAustria
- Department of BiochemistryFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Sadegh Hasannia
- Department of BiochemistryFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Franz Josef Strauss
- Department of Oral BiologyMedical University of ViennaViennaAustria
- Department of Conservative DentistrySchool of DentistryUniversity of ChileSantiagoChile
| | - Reinhard Gruber
- Department of Oral BiologyMedical University of ViennaViennaAustria
- Department of PeriodontologySchool of Dental MedicineUniversity of BernBernSwitzerland
| |
Collapse
|
43
|
Jämsen E, Pajarinen J, Lin TH, Lo CW, Nabeshima A, Lu L, Nathan K, Eklund KK, Yao Z, Goodman SB. Effect of Aging on the Macrophage Response to Titanium Particles. J Orthop Res 2020; 38:405-416. [PMID: 31498470 PMCID: PMC6980287 DOI: 10.1002/jor.24461] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/03/2019] [Indexed: 02/04/2023]
Abstract
Macrophage-mediated inflammatory reaction to implant wear particles drives bone loss around total joint replacements (TJR). Although most TJR recipients are elderly, studies linking wear particle-activated macrophages and peri-implant osteolysis have not taken into account the multiple effects that aging has on the innate immune system and, in particular, on macrophages. To address this, we compared the wear particle responses of bone marrow macrophages obtained from young (2-month) and aged (18-month) mice. Macrophages were polarized to M0, M1, or M2 phenotypes in vitro, challenged with titanium particles, and their inflammatory response was characterized at multiple time points by quantitative reverse-transcription polymerase chain reaction and enzyme-linked immunosorbent assay. In addition, age-dependent changes in activation of transcription factor nuclear factor-κB were analyzed by a lentiviral vector-based luciferase reporter system. The particle stimulation experiment was further repeated using human primary macrophages isolated from blood donors of different ages. We found that the pro-inflammatory responses were generally higher in macrophages obtained from young mice, but differences between the age groups remained small and of uncertain biological significance. Noteworthily, M2 polarization effectively suppressed the particle-induced inflammation in both young and aged macrophages. These results suggest that aging of the innate immune system per se plays no significant role in the response of macrophages to titanium particles, whereas induction of M2 polarization appears a promising strategy to limit macrophage-mediated inflammation regardless of age. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:405-416, 2020.
Collapse
Affiliation(s)
- Eemeli Jämsen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA,Department of Medicine, Clinicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland,Translational Immunology Research Program, University of Helsinki,ORTON Orthopaedic Hospital of the Orton Foundation, Helsinki, Finland
| | - Jukka Pajarinen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA,Department of Medicine, Clinicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland,Translational Immunology Research Program, University of Helsinki,ORTON Orthopaedic Hospital of the Orton Foundation, Helsinki, Finland
| | - Tzu-hua Lin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Chi-Wen Lo
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Akira Nabeshima
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Lu
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Karthik Nathan
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kari K. Eklund
- Department of Medicine, Clinicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland,Translational Immunology Research Program, University of Helsinki,ORTON Orthopaedic Hospital of the Orton Foundation, Helsinki, Finland,University of Helsinki and Helsinki University Hospital, Rheumatology, Helsinki, Finland
| | - Zhenyu Yao
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Stuart B. Goodman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA,Department of Bioengineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
44
|
Tempo-spatial alternative polyadenylation analysis reveals that 3' UTR lengthening of Mdm2 regulates p53 expression and cellular senescence in aged rat testis. Biochem Biophys Res Commun 2020; 523:1046-1052. [PMID: 31973811 DOI: 10.1016/j.bbrc.2020.01.061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/11/2020] [Indexed: 12/21/2022]
Abstract
Although tissue aging is accompanied with cellular senescence, it is much complicated than senescence given both types and number of cells change with age. Alternative polyadenylation (APA) had shown tissue specificity and APA-mediated 3' untranslated region (3' UTR) lengthening could regulate senescence-associated phenotypes. However, whether tissue aging shows similar trends remains unknown. Here, we performed a comprehensive analysis on RNA-seq datasets derived from multiple cells and rat tissues of young and old age. Although APA-mediated 3' UTR lengthening in various senescent cells reinforced the previous discovery, tissue aging showed much more complexity in APA. Interestingly, testis was the only tissue displaying dramatic 3' UTR lengthening and decreased expression trend of corresponding genes in aged rat. Genes with longer 3' UTR in aged testis were enriched in senescence-associated pathways, among which, Mdm2, encoding an E3 ligase of p53, favored distal poly(A) site resulting in lengthened 3' UTR and decreased expression. Longer 3' UTR of Mdm2 generated less protein, and decreased Mdm2 expression led to senescence-associated phenotypes along with increased p53 and p21 protein abundance, which could all be reversed by Mdm2 overexpression. Our work revealed complicated APA changes during tissue aging and discovered APA-mediated 3' UTR lengthening of Mdm2 is a hidden layer in regulating the well-known senescence-related p53-p21 signal axis during testis aging, and also has potential implications regarding declined male fertility along aging.
Collapse
|
45
|
Nishida S, Matsumura T, Senokuchi T, Murakami-Nishida S, Ishii N, Morita Y, Yagi Y, Motoshima H, Kondo T, Araki E. Inhibition of inflammation-mediated DPP-4 expression by linagliptin increases M2 macrophages in atherosclerotic lesions. Biochem Biophys Res Commun 2020; 524:8-15. [PMID: 31964532 DOI: 10.1016/j.bbrc.2020.01.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 01/06/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Dipeptidyl peptidase-4 (DPP-4) inhibitors have been reported to suppress atherosclerosis progression in atherosclerotic mouse models through unclear mechanisms. In this study, we investigated the effect of the DPP-4 inhibitor, linagliptin, on macrophage polarization in vitro and in vivo. METHODS Mouse bone marrow macrophages (BMMs) were used in in vitro assays. High fat diet (HFD)-fed Apoe-/- mice were treated orally with linagliptin (10 mg/kg-1•day-1) or a vehicle (water) control. RESULTS In in vitro assays using BMMs, treatment with LPS and IFNγ decreased the mRNA-expression levels of alternatively activated macrophage (M2) markers, and linagliptin treatment prevented these reductions. The mRNA levels of M2 markers and the number of M2 macrophages in the aorta were higher in linagliptin groups than in control groups. Linagliptin decreased the size of atherosclerotic lesions in HFD-fed Apoe-/- mice. Interestingly, inflammatory stimulation increased DPP-4 expression, and linagliptin suppressed these effects in BMMs. Treatment with DPP-4 small-interfering RNA (siRNA) reproduced linagliptin-mediated alteration of M2 polarization. CONCLUSIONS Linagliptin increased M2 macrophage polarization by inhibiting DPP-4 expression and activity. These findings may indicate the beneficial effects of DPP-4 inhibitors on the progression of diabetic macrovascular complications.
Collapse
Affiliation(s)
- Shuhei Nishida
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Matsumura
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| | - Takafumi Senokuchi
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Saiko Murakami-Nishida
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Norio Ishii
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yutaro Morita
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshitaka Yagi
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Motoshima
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tatsuya Kondo
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Eiichi Araki
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Center for Metabolic Regulation of Healthy Aging (CMHA), Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
46
|
Löffler J, Sass FA, Filter S, Rose A, Ellinghaus A, Duda GN, Dienelt A. Compromised Bone Healing in Aged Rats Is Associated With Impaired M2 Macrophage Function. Front Immunol 2019; 10:2443. [PMID: 31681320 PMCID: PMC6813416 DOI: 10.3389/fimmu.2019.02443] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022] Open
Abstract
Fracture repair is initiated by a multitude of immune cells and induction of an inflammatory cascade. Alterations in the early healing response due to an aged adaptive immune system leads to impaired bone repair, delayed healing or even formation of non-union. However, immuno-senescence is not limited to the adaptive immunity, but is also described for macrophages, main effector cells from the innate immune system. Beside regulation of pro- and anti-inflammatory signaling, macrophages contribute to angiogenesis and granulation tissue maturation. Thus, it seems likely that an altered macrophage function due to aging may affect bone repair at various stages and contribute to age related deficiencies in bone regeneration. To prove this hypothesis, we analyzed the expression of macrophage markers and angiogenic factors in the early bone hematoma derived from young and aged osteotomized Spraque Dawley rats. We detected an overall reduced expression of the monocyte/pan-macrophage markers CD14 and CD68 in aged rats. Furthermore, the analysis revealed an impaired expression of anti-inflammatory M2 macrophage markers in hematoma from aged animals that was connected to a diminished revascularization of the bone callus. To verify that the age related disturbed bone regeneration was due to a compromised macrophage function, CD14+ macrophage precursors were transplanted locally into the osteotomy gap of aged rats. Transplantation rescued bone regeneration partially after 6 weeks, demonstrated by a significantly induced deposition of new bone tissue, reduced fibrosis and significantly improved callus vascularization.
Collapse
Affiliation(s)
- Julia Löffler
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - F Andrea Sass
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sebastian Filter
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alexander Rose
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Agnes Ellinghaus
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anke Dienelt
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
47
|
Goodman SB, Pajarinen J, Yao Z, Lin T. Inflammation and Bone Repair: From Particle Disease to Tissue Regeneration. Front Bioeng Biotechnol 2019; 7:230. [PMID: 31608274 PMCID: PMC6761220 DOI: 10.3389/fbioe.2019.00230] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 09/06/2019] [Indexed: 12/17/2022] Open
Abstract
When presented with an adverse stimulus, organisms evoke an immediate, pre-programmed, non-specific innate immune response. The purpose of this reaction is to maintain the organism's biological integrity and function, mitigate or eradicate the injurious source, and re-establish tissue homeostasis. The initial stage of this protective reaction is acute inflammation, which normally reduces or terminates the offending stimulus. As the inflammatory reaction recedes, the stage of tissue repair and regeneration follows. If the above sequence of events is perturbed, reconstitution of normal biological form and function will not be achieved. Dysregulation of these activities may result in incomplete healing, fibrosis, or chronic inflammation. Our laboratory has studied the reaction to wear particles from joint replacements as a paradigm for understanding the biological pathways of acute and chronic inflammation, and potential translational treatments to reconstitute lost bone. As inflammation is the cornerstone for healing in all anatomical locations, the concepts developed have relevance to tissue engineering and regenerative medicine in all organ systems. To accomplish our goal, we developed novel in vitro and in vivo models (including the murine femoral continuous intramedullary particle infusion model), translational strategies including modulation of macrophage chemotaxis and polarization, and methods to interfere with key transcription factors NFκB and MyD88. We purposefully modified MSCs to facilitate bone healing in inflammatory scenarios: by preconditioning the MSCs, and by genetically modifying MSCs to first sense NFκB activation and then overexpress the anti-inflammatory pro-regenerative cytokine IL-4. These advancements provide significant translational opportunities to enhance healing in bone and other organs.
Collapse
Affiliation(s)
- Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Redwood City, CA, United States.,Department of Bioengineering, Stanford University, Stanford, CA, United States.,Department of Medicine, Clinicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jukka Pajarinen
- Department of Medicine, Clinicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Zhenyu Yao
- Orthopaedic Research Laboratories, Stanford University, Stanford, CA, United States
| | - Tzuhua Lin
- Orthopaedic Research Laboratories, Stanford University, Stanford, CA, United States
| |
Collapse
|
48
|
Sequential drug delivery to modulate macrophage behavior and enhance implant integration. Adv Drug Deliv Rev 2019; 149-150:85-94. [PMID: 31103451 DOI: 10.1016/j.addr.2019.05.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/03/2019] [Accepted: 05/15/2019] [Indexed: 12/30/2022]
Abstract
Macrophages are major upstream regulators of the inflammatory response to implanted biomaterials. Sequential functions of distinct macrophage phenotypes are essential to the normal tissue repair process, which ideally results in vascularization and integration of implants. Improper timing of M1 or M2 macrophage activation results in dysfunctional healing in the form of chronic inflammation or fibrous encapsulation of the implant. Thus, biphasic drug delivery systems that modulate macrophage behavior are an appealing approach to promoting implant integration. In this review, we describe the timing and roles of macrophage phenotypes in healing, then highlight current drug delivery systems designed to sequentially modulate macrophage behavior.
Collapse
|
49
|
Ghrelin Signaling in Immunometabolism and Inflamm-Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1090:165-182. [PMID: 30390290 DOI: 10.1007/978-981-13-1286-1_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intracellular changes in immune cells lead to metabolic dysfunction, which is termed immunometabolism. Chronic inflammation is a hallmark of aging; this phenomenon is described as inflamm-aging. Immunometabolism and inflamm-aging are closely linked to obesity, insulin resistance, type 2 diabetes (T2D), cardiovascular diseases, and cancers, which consequently reduce life span and health span of the elderly. Ghrelin is an orexigenic hormone that regulates appetite and food intake. Ghrelin's functions are mediated through its receptor, growth hormone secretagogue receptor (GHS-R). Ghrelin and GHS-R have important roles in age-associated obesity, insulin resistance, and T2D. In this chapter, we have discussed the roles of ghrelin signaling in diet-induced obesity and normal aging as it relates to energy metabolism and inflammation in key metabolic tissues and organs. The new findings reveal that ghrelin signaling is an important regulatory mechanism for immunometabolism and inflamm-aging. Ghrelin signaling offers an exciting novel therapeutic strategy for treatment of obesity and insulin resistance of the elderly.
Collapse
|
50
|
Bucher CH, Schlundt C, Wulsten D, Sass FA, Wendler S, Ellinghaus A, Thiele T, Seemann R, Willie BM, Volk HD, Duda GN, Schmidt-Bleek K. Experience in the Adaptive Immunity Impacts Bone Homeostasis, Remodeling, and Healing. Front Immunol 2019; 10:797. [PMID: 31031773 PMCID: PMC6474158 DOI: 10.3389/fimmu.2019.00797] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Abstract
Bone formation as well as bone healing capacity is known to be impaired in the elderly. Although bone formation is outpaced by bone resorption in aged individuals, we hereby present a novel path that considerably impacts bone formation and architecture: Bone formation is substantially reduced in aged individual owing to the experience of the adaptive immunity. Thus, immune-aging in addition to chronological aging is a potential risk factor, with an experienced immune system being recognized as more pro-inflammatory. The role of the aging immune system on bone homeostasis and on the bone healing cascade has so far not been considered. Within this study mice at different age and immunological experience were analyzed toward bone properties. Healing was assessed by introducing an osteotomy, immune cells were adoptively transferred to disclose the difference in biological vs. chronological aging. In vitro studies were employed to test the interaction of immune cell products (cytokines) on cells of the musculoskeletal system. In metaphyseal bone, immune-aging affects bone homeostasis by impacting bone formation capacity and thereby influencing mass and microstructure of bone trabeculae leading to an overall reduced mechanical competence as found in bone torsional testing. Furthermore, bone formation is also impacted during bone regeneration in terms of a diminished healing capacity observed in young animals who have an experienced human immune system. We show the impact of an experienced immune system compared to a naïve immune system, demonstrating the substantial differences in the healing capacity and bone homeostasis due to the immune composition. We further showed that in vivo mechanical stimulation changed the immune system phenotype in young mice toward a more naïve composition. While this rescue was found to be significant in young individuals, aged mice only showed a trend toward the reconstitution of a more naïve immune phenotype. Considering the immune system's experience level in an individual, will likely allow one to differentiate (stratify) and treat (immune-modulate) patients more effectively. This work illustrates the relevance of including immune diagnostics when discussing immunomodulatory therapeutic strategies for the progressively aging population of the industrial countries.
Collapse
Affiliation(s)
- Christian H Bucher
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Schlundt
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dag Wulsten
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - F Andrea Sass
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Wendler
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Agnes Ellinghaus
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Thiele
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ricarda Seemann
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bettina M Willie
- Department of Pediatric Surgery, Faculty of Medicine, McGill University, Shriners Hospital for Children, Montreal, QC, Canada
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|