1
|
Tjandra PM, Orr SV, Lam SK, Kulkarni AD, Chen YJ, Adhikari A, Silverman JL, Ripplinger CM, Christiansen BA. Investigating the role of complement 5a in systemic bone loss after myocardial infarction. Bone 2025:117543. [PMID: 40412474 DOI: 10.1016/j.bone.2025.117543] [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: 01/31/2025] [Revised: 05/13/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
Myocardial infarction (MI) and osteoporotic fracture are two of the leading causes of morbidity and mortality worldwide. We have previously established that MI in mice directly causes post-traumatic systemic bone loss and that the sympathetic nervous system plays a role in this response. However, the systemic injury response is mediated by multiple systems. In this study, we investigated the role of complement 5a (C5a), one of the main mediators driving multiple organ dysfunction after trauma. MI was surgically induced in 12-week-old male C57BL/6 J mice (B6/J), C5a receptor 1 knockout mice (C5aR1-/-) and B10·D2-Hc0 H2d H2-T18c/oSnJ (B10·D2), (JAX Strain # 000461) mice that are deficient in serum C5. Systemic and localized bone changes were analyzed at 7-, 14- and 28-days post-MI using micro-computed tomography and three-point bending mechanical testing. Osteoclast number and activity was quantified using tartrate-resistant acid phosphatase (TRAP) staining, and voluntary activity levels were measured using open field. We found that MI induced peak trabecular bone loss 7 days after injury in the L5 vertebral body and caused reductions in femoral cortical bone 28 days post-MI. However, MI did not impact femoral trabecular bone in this timeframe. B10·D2 mice had reduced trabecular and cortical bone morphology compared to B6 and C5aR1-/- mice, but did not exhibit an altered response to MI. Osteoclast activity 7-days post-MI was increased in C5aR1-/- mice compared to B6 and B10·D2 mice, but MI did not impact osteoclast activity at this time point. Altogether, these findings suggest that C5a may influence overall response to MI and bone morphology instead of post-traumatic systemic bone loss response following MI, though likely not as a primary mechanism.
Collapse
Affiliation(s)
- Priscilla M Tjandra
- University of California Davis, Department of Orthopaedic Surgery, United States of America
| | - Sophie V Orr
- University of California Davis, Department of Orthopaedic Surgery, United States of America
| | - Selena K Lam
- University of California Davis, Department of Orthopaedic Surgery, United States of America
| | - Anika D Kulkarni
- University of California Davis, Department of Orthopaedic Surgery, United States of America
| | - Yi-Je Chen
- University of California Davis, Department of Pharmacology, United States of America
| | - Anna Adhikari
- University of California Davis, Department of Psychiatry and Behavioral Sciences and the MIND Institute, University of California Davis School of Medicine, United States of America
| | - Jill L Silverman
- University of California Davis, Department of Psychiatry and Behavioral Sciences and the MIND Institute, University of California Davis School of Medicine, United States of America
| | - Crystal M Ripplinger
- University of California Davis, Department of Pharmacology, United States of America
| | - Blaine A Christiansen
- University of California Davis, Department of Orthopaedic Surgery, United States of America.
| |
Collapse
|
2
|
Carroll KA, Sawden M, Sharma S. DAMPs, PAMPs, NLRs, RIGs, CLRs and TLRs - Understanding the Alphabet Soup in the Context of Bone Biology. Curr Osteoporos Rep 2025; 23:6. [PMID: 39808398 DOI: 10.1007/s11914-024-00900-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the current understanding of cell-autonomous innate immune pathways that contribute to bone homeostasis and disease. RECENT FINDINGS Germ-line encoded pattern recognition receptors (PRRs) are the first line of defense against danger and infections. In the bone microenvironment, PRRs and downstream signaling pathways, that mount immune defense, interface intimately with the core cellular processes in bone cells to alter bone formation and resorption. The role of PRR engagement on bone remodeling has been best described as a result of activated macrophages secreting effector molecules that reshape the characteristics of bone-resident cells. However, it is being increasingly recognized that local bone resident-cells like osteoclasts and osteoblasts possess an arsenal of PRRs. The engagement of these PRRs by stimuli in the bone niche can drive cell-autonomous (aka cell-intrinsic) responses that, in turn, impact bone-remodeling dramatically, irrespective of immune cell effectors. Indeed, this vital role for cell-autonomous innate immune responses is evident in how reduced PRR activity within osteoclast progenitors correlates with their reduced differentiation and abnormal bone remodeling. Further, cell-intrinsic PRR activity has now been shown to influence the behavior of osteoblasts, osteocytes and other local immune/non-immune cell populations. However, distinct PRR families have varying impact on bone homeostasis and inflammation, emphasizing the importance of investigating these different nodes of innate immune signaling in bone cells to better identify how they synergistically and/or antagonistically regulate bone remodeling in the course of an immune response. Innate immune sensing within bone resident cells is a critical determinant for bone remodeling in health and disease.
Collapse
Affiliation(s)
- K A Carroll
- Department of Immunology, Tufts University, Boston, MA, 02111, USA
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
| | - M Sawden
- Department of Immunology, Tufts University, Boston, MA, 02111, USA
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
| | - S Sharma
- Department of Immunology, Tufts University, Boston, MA, 02111, USA.
| |
Collapse
|
3
|
Ruths L, Huber-Lang M, Schulze-Tanzil G, Riegger J. Anaphylatoxins and their corresponding receptors as potential drivers in cartilage calcification during osteoarthritis progression. Osteoarthritis Cartilage 2024; 32:514-525. [PMID: 38242312 DOI: 10.1016/j.joca.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/21/2024]
Abstract
OBJECTIVE The complement cascade as major fluid phase innate immune system is activated during progression of osteoarthritis (OA). Generated anaphylatoxins and the corresponding receptors C3aR and C5aR1 are associated with the calcification of blood vessels and involved in osteogenic differentiation. This study aims on elucidating whether complement activation products contribute to cartilage calcification of OA cartilage. METHOD Human articular chondrocytes were osteogenically differentiated in vitro in the presence or absence of C3a, C5a, and bone morphogenetic protein (BMP) 2. Furthermore, macroscopically intact (OARSI grade ≤ 1) and highly degenerated human cartilage (OARSI grade ≥ 3) was used for C3aR and C5aR1 histochemistry. Calcification of the cartilage was assessed by Alizarin Red S and von Kossa staining. RESULTS C3a and C5a amplified matrix mineralization during in vitro osteogenesis, while inhibition of the corresponding receptors impaired calcium deposition. Moreover, C3aR and C5aR1 expression was upregulated during osteogenic differentiation and also in degenerated cartilage. Additionally, anaphylatoxin receptor expression was positively associated with calcification of native cartilage tissue and calcium deposition during osteogenic differentiation. Finally, the pro-hypertrophic growth factor BMP2 induced the expression of C5aR1. CONCLUSIONS Our findings indicate that anaphylatoxins and their receptors play a decisive role in cartilage calcification processes during OA progression.
Collapse
Affiliation(s)
- Leonie Ruths
- Division for Biochemistry of Joint and Connective Tissue Diseases, University Hospital Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Gundula Schulze-Tanzil
- Department of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg, Germany
| | - Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
4
|
Chen W, Wang Q, Tao H, Lu L, Zhou J, Wang Q, Huang W, Yang X. Subchondral osteoclasts and osteoarthritis: new insights and potential therapeutic avenues. Acta Biochim Biophys Sin (Shanghai) 2024; 56:499-512. [PMID: 38439665 DOI: 10.3724/abbs.2024017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is the most common joint disease, and good therapeutic results are often difficult to obtain due to its complex pathogenesis and diverse causative factors. After decades of research and exploration of OA, it has been progressively found that subchondral bone is essential for its pathogenesis, and pathological changes in subchondral bone can be observed even before cartilage lesions develop. Osteoclasts, the main cells regulating bone resorption, play a crucial role in the pathogenesis of subchondral bone. Subchondral osteoclasts regulate the homeostasis of subchondral bone through the secretion of degradative enzymes, immunomodulation, and cell signaling pathways. In OA, osteoclasts are overactivated by autophagy, ncRNAs, and Rankl/Rank/OPG signaling pathways. Excessive bone resorption disrupts the balance of bone remodeling, leading to increased subchondral bone loss, decreased bone mineral density and consequent structural damage to articular cartilage and joint pain. With increased understanding of bone biology and targeted therapies, researchers have found that the activity and function of subchondral osteoclasts are affected by multiple pathways. In this review, we summarize the roles and mechanisms of subchondral osteoclasts in OA, enumerate the latest advances in subchondral osteoclast-targeted therapy for OA, and look forward to the future trends of subchondral osteoclast-targeted therapies in clinical applications to fill the gaps in the current knowledge of OA treatment and to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Wenlong Chen
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Qiufei Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Huaqiang Tao
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Lingfeng Lu
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Jing Zhou
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Qiang Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Wei Huang
- Department of Orthopaedics, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| |
Collapse
|
5
|
Tjandra PM, Ripplinger CM, Christiansen BA. The heart-bone connection: relationships between myocardial infarction and osteoporotic fracture. Am J Physiol Heart Circ Physiol 2024; 326:H845-H856. [PMID: 38305753 PMCID: PMC11062618 DOI: 10.1152/ajpheart.00576.2023] [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: 09/18/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024]
Abstract
Myocardial infarction (MI) and osteoporotic fracture (Fx) are two of the leading causes of mortality and morbidity worldwide. Although these traumatic injuries are treated as if they are independent, there is epidemiological evidence linking the incidence of Fx and MI, thus raising the question of whether each of these events can actively influence the risk of the other. Atherosclerotic cardiovascular disease and osteoporosis, the chronic conditions leading to MI and Fx, are known to have shared pathoetiology. Furthermore, sustained systemic inflammation after traumas such as MI and Fx has been shown to exacerbate both underlying chronic conditions. However, the effects of MI and Fx outside their own system have not been well studied. The sympathetic nervous system (SNS) and the complement system initiate a systemic response after MI that could lead to subsequent changes in bone remodeling through osteoclasts. Similarly, SNS and complement system activation following fracture could lead to heart tissue damage and exacerbate atherosclerosis. To determine whether damaging bone-heart cross talk may be important comorbidity following Fx or MI, this review details the current understanding of bone loss after MI, cardiovascular damage after Fx, and possible shared underlying mechanisms of these processes.
Collapse
Affiliation(s)
- Priscilla M Tjandra
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, United States
- Biomedical Engineering Graduate Group, University of California Davis, Davis, California, United States
| | - Crystal M Ripplinger
- Biomedical Engineering Graduate Group, University of California Davis, Davis, California, United States
- Department of Pharmacology, University of California Davis Health, Davis, California, United States
| | - Blaine A Christiansen
- Biomedical Engineering Graduate Group, University of California Davis, Davis, California, United States
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, California, United States
| |
Collapse
|
6
|
Yu S, Lv L, Li Y, Ning Q, Liu T, Hu T. PLK3 promotes the proneural-mesenchymal transition in glioblastoma via transcriptional regulation of C5AR1. Mol Biol Rep 2023; 50:8249-8258. [PMID: 37568042 DOI: 10.1007/s11033-023-08716-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023]
Abstract
BACKGROUND Accumulating evidence suggests that polo-like kinase 3 (PLK3) plays an essential role in tumor cells and induces cell proliferation and may have implications for the prognosis of various cancers. We sought to define the role of PLK3-dependent proneural-mesenchymal transition (PMT) in the glioblastoma (GBM) therapy. METHODS AND RESULTS We analyzed the expression data for PLK3 by using the TCGA database. PLK3 expression in GBM cell lines was determined by qRT-PCR and Western blotting. PLK3 levels were modulated using Lentivirus infection, and the effects on symptoms, tumor volume, and survival in mice intracranial xenograft models were determined. Irradiation (IR) was performed to induce PMT. PLK3 expression was significantly elevated in mesenchymal subtype GBM and promoted tumor proliferation in GBM. Additionally enriched PLK3 expression could be associated with poor prognosis in GBM patients compared with those who have lower PLK3 expression. Mechanically, PLK3-dependent PMT induced radioresistance in GBM cells via transcriptional regulation of complement C5a receptor 1 (C5AR1). In therapeutic experiments conducted in vitro, targeting PLK3 by using small molecule inhibitor decreased tumor growth and radioresistance of GBM cells both in vitro and in vivo. CONCLUSIONS PLK3-C5AR1 axis induced PMT thus enhanced radioresistance in GBM and could become a novel potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Shuo Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, Shaanxi, China
| | - Lin Lv
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China
| | - Yang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China
| | - Qian Ning
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China
| | - Tingting Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China
| | - Tinghua Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710000, Shaanxi, China.
| |
Collapse
|
7
|
Kuhn MB, VandenBerg HS, Reynolds AJ, Carson MD, Warner AJ, LaRue AC, Novince CM, Hathaway-Schrader JD. C3a-C3aR signaling is a novel modulator of skeletal homeostasis. Bone Rep 2023; 18:101662. [PMID: 36860797 PMCID: PMC9969257 DOI: 10.1016/j.bonr.2023.101662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
Osteoimmune studies have identified complement signaling as an important regulator of the skeleton. Specifically, complement anaphylatoxin receptors (i.e., C3aR, C5aR) are expressed on osteoblasts and osteoclasts, implying that C3a and/or C5a may be candidate mediators of skeletal homeostasis. The study aimed to determine how complement signaling influences bone modeling/remodeling in the young skeleton. Female C57BL/6J C3aR-/-C5aR-/- vs. wildtype and C3aR-/- vs. wildtype mice were examined at age 10 weeks. Trabecular and cortical bone parameters were analyzed by micro-CT. In situ osteoblast and osteoclast outcomes were determined by histomorphometry. Osteoblast and osteoclast precursors were assessed in vitro. C3aR-/-C5aR-/- mice displayed an increased trabecular bone phenotype at age 10 weeks. In vitro studies revealed C3aR-/-C5aR-/- vs. wildtype cultures had less bone-resorbing osteoclasts and increased bone-forming osteoblasts, which were validated in vivo. To determine whether C3aR alone was critical for the enhanced skeletal outcomes, wildtype vs. C3aR-/- mice were evaluated for osseous tissue outcomes. Paralleling skeletal findings in C3aR-/-C5aR-/- mice, C3aR-/- vs. wildtype mice had an enhanced trabecular bone volume fraction, which was attributed to increased trabecular number. There was elevated osteoblast activity and suppressed osteoclastic cells in C3aR-/- vs. wildtype mice. Furthermore, primary osteoblasts derived from wildtype mice were stimulated with exogenous C3a, which more profoundly upregulated C3ar1 and the pro-osteoclastic chemokine Cxcl1. This study introduces the C3a/C3aR signaling axis as a novel regulator of the young skeleton.
Collapse
Affiliation(s)
- Megan B. Kuhn
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Hayden S. VandenBerg
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Andrew J. Reynolds
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Matthew D. Carson
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Stomatology-Div. of Periodontics, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Pediatrics-Div. of Endocrinology, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Amy J. Warner
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Stomatology-Div. of Periodontics, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Pediatrics-Div. of Endocrinology, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Amanda C. LaRue
- Research Services, Ralph H. Johnson Department of Veterans Affairs Health Care System, Charleston, SC, USA
- Department of Pathology and Laboratory Medicine, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Chad M. Novince
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Stomatology-Div. of Periodontics, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Pediatrics-Div. of Endocrinology, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Jessica D. Hathaway-Schrader
- Department of Stomatology-Div. of Periodontics, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
- Research Services, Ralph H. Johnson Department of Veterans Affairs Health Care System, Charleston, SC, USA
- Department of Pathology and Laboratory Medicine, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
8
|
Haffner-Luntzer M, Weber B, Morioka K, Lackner I, Fischer V, Bahney C, Ignatius A, Kalbitz M, Marcucio R, Miclau T. Altered early immune response after fracture and traumatic brain injury. Front Immunol 2023; 14:1074207. [PMID: 36761764 PMCID: PMC9905106 DOI: 10.3389/fimmu.2023.1074207] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
Introduction Clinical and preclinical data suggest accelerated bone fracture healing in subjects with an additional traumatic brain injury (TBI). Mechanistically, altered metabolism and neuro-endocrine regulations have been shown to influence bone formation after combined fracture and TBI, thereby increasing the bone content in the fracture callus. However, the early inflammatory response towards fracture and TBI has not been investigated in detail so far. This is of great importance, since the early inflammatory phase of fracture healing is known to be essential for the initiation of downstream regenerative processes for adequate fracture repair. Methods Therefore, we analyzed systemic and local inflammatory mediators and immune cells in mice which were exposed to fracture only or fracture + TBI 6h and 24h after injury. Results We found a dysregulated systemic immune response and significantly fewer neutrophils and mast cells locally in the fracture hematoma. Further, local CXCL10 expression was significantly decreased in the animals with combined trauma, which correlated significantly with the reduced mast cell numbers. Discussion Since mast cells and mast cell-derived CXCL10 have been shown to increase osteoclastogenesis, the reduced mast cell numbers might contribute to higher bone content in the fracture callus of fracture + TBI mice due to decreased callus remodeling.
Collapse
Affiliation(s)
- Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany.,Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Birte Weber
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany.,Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Kazuhito Morioka
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Ina Lackner
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Chelsea Bahney
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States.,Steadman Phillipon Research Institute, Vail, CO, United States
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
9
|
Shirazi S, Ravindran S, Cooper LF. Topography-mediated immunomodulation in osseointegration; Ally or Enemy. Biomaterials 2022; 291:121903. [PMID: 36410109 PMCID: PMC10148651 DOI: 10.1016/j.biomaterials.2022.121903] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Osteoimmunology is at full display during endosseous implant osseointegration. Bone formation, maintenance and resorption at the implant surface is a result of bidirectional and dynamic reciprocal communication between the bone and immune cells that extends beyond the well-defined osteoblast-osteoclast signaling. Implant surface topography informs adherent progenitor and immune cell function and their cross-talk to modulate the process of bone accrual. Integrating titanium surface engineering with the principles of immunology is utilized to harness the power of immune system to improve osseointegration in healthy and diseased microenvironments. This review summarizes current information regarding immune cell-titanium implant surface interactions and places these events in the context of surface-mediated immunomodulation and bone regeneration. A mechanistic approach is directed in demonstrating the central role of osteoimmunology in the process of osseointegration and exploring how regulation of immune cell function at the implant-bone interface may be used in future control of clinical therapies. The process of peri-implant bone loss is also informed by immunomodulation at the implant surface. How surface topography is exploited to prevent osteoclastogenesis is considered herein with respect to peri-implant inflammation, osteoclastic precursor-surface interactions, and the upstream/downstream effects of surface topography on immune and progenitor cell function.
Collapse
Affiliation(s)
- Sajjad Shirazi
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA.
| | - Sriram Ravindran
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA
| | - Lyndon F Cooper
- School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
10
|
Ruocco A, Sirico A, Novelli R, Iannelli S, Van Breda SV, Kyburz D, Hasler P, Aramini A, Amendola PG. The role of C5a-C5aR1 axis in bone pathophysiology: A mini-review. Front Cell Dev Biol 2022; 10:957800. [PMID: 36003145 PMCID: PMC9393612 DOI: 10.3389/fcell.2022.957800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Bone remodeling is a physiological, dynamic process that mainly depends on the functions of 2 cell types: osteoblasts and osteoclasts. Emerging evidence suggests that complement system is crucially involved in the regulation of functions of these cells, especially during inflammatory states. In this context, complement component 5a (C5a), a powerful pro-inflammatory anaphylatoxin that binds the receptor C5aR1, is known to regulate osteoclast formation and osteoblast inflammatory responses, and has thus been proposed as potential therapeutic target for the treatment of inflammatory bone diseases. In this review, we will analyze the role of C5a-C5aR1 axis in bone physiology and pathophysiology, describing its involvement in the pathogenesis of some of the most frequent inflammatory bone diseases such as rheumatoid arthritis, and also in osteoporosis and bone cancer and metastasis. Moreover, we will examine C5aR1-based pharmacological approaches that are available and have been tested so far for the treatment of these conditions. Given the growing interest of the scientific community on osteoimmunology, and the scarcity of data regarding the role of C5a-C5aR1 axis in bone pathophysiology, we will highlight the importance of this axis in mediating the interactions between skeletal and immune systems and its potential use as a therapeutic target.
Collapse
Affiliation(s)
| | | | | | | | | | - Diego Kyburz
- Departement Biomedizin, University of Basel, Basel, Switzerland
| | - Paul Hasler
- Division of Rheumatology, Kantonsspital Aarau AG, Aarau, Switzerland
| | | | | |
Collapse
|
11
|
Schäfer N, Grässel S. Involvement of complement peptides C3a and C5a in osteoarthritis pathology. Peptides 2022; 154:170815. [PMID: 35598724 DOI: 10.1016/j.peptides.2022.170815] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 12/28/2022]
Abstract
Osteoarthritis (OA) affects more than 500 million people worldwide and is among the five diseases in Germany causing the highest suffering of the patients and cost for the society. The quality of life of OA patients is severely compromised, and adequate therapy is lacking owing to a knowledge gap that acts as a major barrier to finding safe and effective solutions. Chronic, low-grade inflammation plays a central role in OA pathogenesis and is associated with both OA pain and disease progression. Innate immune pathways, such as the complement- and pattern-recognition receptor pathways, are pivotal to the inflammation in OA and key components of the innate immune system implicated in OA include DAMP-TLR signaling, the complement system, carboxypeptidase B (CPB), and mononuclear cells. Anaphylatoxins C3a and C5a are small polypeptides (77 and 74 amino acids, respectively) which are released by proteolytic cleavage of the complement components C3 and C5. The alternative complement pathway seems to play a crucial role in OA pathogenesis as these complement components, mostly C3 and its activation peptide C3a, were detected at high levels in osteoarthritic cartilage, synovial membrane, and cultured chondrocytes. Targeting the complement system by using anti-complement drugs as a therapeutic option bears the risk of major side effects such as increasing the risk of infection, interfering with cell regeneration and metabolism, and suppressing the clearance of immune complexes. Despite those adverse effects, several synthetic complement peptide antagonists show promising effects in ameliorating inflammatory cell responses also in joint tissues.
Collapse
Affiliation(s)
- Nicole Schäfer
- Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), Bio Park 1, University of Regensburg, Germany
| | - Susanne Grässel
- Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), Bio Park 1, University of Regensburg, Germany; Department of Orthopaedic Surgery, University of Regensburg, Germany.
| |
Collapse
|
12
|
Ragipoglu D, Bülow J, Hauff K, Voss M, Haffner-Luntzer M, Dudeck A, Ignatius A, Fischer V. Mast Cells Drive Systemic Inflammation and Compromised Bone Repair After Trauma. Front Immunol 2022; 13:883707. [PMID: 35558068 PMCID: PMC9086903 DOI: 10.3389/fimmu.2022.883707] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
There is evidence that mast cells contribute to inflammation induced by hemorrhagic shock, severe tissue injury or sepsis. Mast cells are highly responsive to alarm signals generated after trauma, and release many inflammatory mediators including interleukin-6, a key mediator of posttraumatic inflammation. An overwhelming posttraumatic inflammation causes compromised bone healing; however, the underlying cellular and molecular mechanisms are poorly understood. Recently, we found that mast cells trigger local and systemic inflammation after isolated fracture leading to uneventful bone repair. Here, we investigated whether mast cells critically contribute to trauma-induced compromised bone healing. Male Mcpt5-Cre+ R-DTA mice, which lack connective tissue type mast cells, and their mast cell-competent Cre- littermates underwent a femur fracture with/without thoracic trauma. Posttraumatic systemic and local inflammation and bone repair were assessed 3 h and 21 d post injury. Both, the systemic and pulmonary inflammation was significantly increased in mast cell-competent mice upon combined trauma compared to isolated fracture. In mast cell-deficient mice, the increase of inflammatory mediators in the circulation induced by the severe trauma was abolished. In the bronchoalveolar lavage fluid, the trauma-induced increase of inflammatory cytokines was not reduced, but the neutrophil invasion into the lungs was significantly diminished in the absence of mast cells. Locally in the fracture hematoma, mast cell-competent mice displayed reduced inflammatory mediator concentrations after combined trauma compared to isolated fracture, which was abolished in mast cell-deficient mice. Notably, while combined trauma resulted in compromised bone repair in mast cell-competent mice, indicated by significantly reduced bone and increased cartilage fracture callus contents, this was abolished in Mcpt5-Cre+ R-DTA mice. Therefore, mast cells contribute to trauma-induced compromised bone repair and could be a potential target for new treatment options to improve fracture healing in multiply injured patients.
Collapse
Affiliation(s)
- Deniz Ragipoglu
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Jasmin Bülow
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Kristin Hauff
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Martin Voss
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Anne Dudeck
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
13
|
Bülow JM, Renz N, Haffner-Luntzer M, Fischer V, Schoppa A, Tuckermann J, Köhl J, Huber-Lang M, Ignatius A. Complement receptor C5aR1 on osteoblasts regulates osteoclastogenesis in experimental postmenopausal osteoporosis. Front Endocrinol (Lausanne) 2022; 13:1016057. [PMID: 36246887 PMCID: PMC9561253 DOI: 10.3389/fendo.2022.1016057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
In recent years, evidence has accumulated that the complement system, an integral part of innate immunity, may be involved in the regulation of bone homeostasis as well as inflammatory bone loss, for example, in rheumatoid arthritis and periodontitis. Complement may also contribute to osteoporosis development, but investigation of the mechanism is limited. Using mice with a conditional deletion of the complement anaphylatoxin receptor C5aR1, we here demonstrated that C5aR1 in osteoblasts (C5aR1 Runx2-Cre mice) or osteoclasts (C5aR1 LysM-Cre mice) did not affect physiological bone turnover or age-related bone loss in either sex, as confirmed by micro-computed tomography, histomorphometry, and biomechanical analyses of the bone and by the measurement of bone turnover markers in the blood serum. When female mice were subjected to ovariectomy (OVX), a common model for postmenopausal osteoporosis, significant bone loss was induced in C5aR1 fl/fl and C5aR1 LysM-Cre mice, as demonstrated by a significantly reduced bone volume fraction, trabecular number and thickness as well as an increased trabecular separation in the trabecular bone compartment. Confirming this, the osteoclast number and the receptor activator of nuclear factor k-B (RANK) ligand (RANKL) serum level were significantly elevated in these mouse lines. By contrast, C5aR1 Runx2-Cre mice were protected from bone loss after OVX and the serum RANKL concentration was not increased after OVX. These data suggested that bone cell-specific C5aR1 may be redundant in bone homeostasis regulation under physiological conditions. However, C5aR1 on osteoblasts was crucial for the induction of bone resorption under osteoporotic conditions by stimulating RANKL release, whereas C5aR1 on osteoclasts did not regulate OVX-induced bone loss. Therefore, our results implicate C5aR1 on osteoblasts as a potential target for treating postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Jasmin Maria Bülow
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Nikolai Renz
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University Medical Center, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
- *Correspondence: Anita Ignatius,
| |
Collapse
|
14
|
Fischer V, Ragipoglu D, Diedrich J, Steppe L, Dudeck A, Schütze K, Kalbitz M, Gebhard F, Haffner-Luntzer M, Ignatius A. Mast Cells Trigger Disturbed Bone Healing in Osteoporotic Mice. J Bone Miner Res 2022; 37:137-151. [PMID: 34633111 DOI: 10.1002/jbmr.4455] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/21/2021] [Accepted: 10/03/2021] [Indexed: 12/16/2022]
Abstract
Mast cells are important tissue-resident sensor and effector immune cells but also play a major role in osteoporosis development. Mast cells are increased in numbers in the bone marrow of postmenopausal osteoporotic patients, and mast cell-deficient mice are protected from ovariectomy (OVX)-induced bone loss. In this study, we showed that mast cell-deficient Mcpt5-Cre R-DTA mice were protected from OVX-induced disturbed fracture healing, indicating a critical role for mast cells in the pathomechanisms of impaired bone repair under estrogen-deficient conditions. We revealed that mast cells trigger the fracture-induced inflammatory response by releasing inflammatory mediators, including interleukin-6, midkine (Mdk), and C-X-C motif chemokine ligand 10 (CXCL10), and promote neutrophil infiltration into the fracture site in OVX mice. Furthermore, mast cells were responsible for reduced osteoblast and increased osteoclast activities in OVX mice callus, as well as increased receptor activator of NF-κB ligand serum levels in OVX mice. Additional in vitro studies with human cells showed that mast cells stimulate osteoclastogenesis by releasing the osteoclastogenic mediators Mdk and CXCL10 in an estrogen-dependent manner, which was mediated via the estrogen receptor alpha on mast cells. In conclusion, mast cells negatively affect the healing of bone fractures under estrogen-deficient conditions. Hence, targeting mast cells might provide a therapeutic strategy to improve disturbed bone repair in postmenopausal osteoporosis. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Verena Fischer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Deniz Ragipoglu
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Johanna Diedrich
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Lena Steppe
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Anne Dudeck
- Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Konrad Schütze
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany.,Department of Trauma and Orthopedic Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen-Nürnberg, Germany
| | - Florian Gebhard
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
15
|
Zheng JM, Zhou HX, Yu HY, Xia YH, Yu QX, Qu HS, Bao JQ. By Increasing the Expression and Activation of STAT3, Sustained C5a Stimulation Increases the Proliferation, Migration, and Invasion of RCC Cells and Promotes the Growth of Transgrafted Tumors. Cancer Manag Res 2021; 13:7607-7621. [PMID: 34675657 PMCID: PMC8500505 DOI: 10.2147/cmar.s326352] [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: 06/24/2021] [Accepted: 09/11/2021] [Indexed: 11/23/2022] Open
Abstract
Background Contradictive results about the direct role of C5a/C5aR1 axis in different cancer cells have been reported. The direct effect of C5a on human renal cell carcinoma (RCC) cells and the underlying mechanism are not clear. The aim of this study is to investigate the role of C5a/C5aR1 axis in RCC cells and its working mechanism. Methods RCC cells were infected with lentivirus Lenti-C5a, which was designed to over-express secretory C5a in the cells, or directly treated with recombinant C5a, the influence of these treatments in the cells and the underlying mechanism were explored. Results Transfection of RCC cells with Lenti-C5a markedly increased the production of C5a and significantly increased the proliferation, migration, and invasion of RCC cells, but direct addition of C5a to the cell culture medium had no such effects though it indeed induced a transient intracellular calcium rise. RCC cells were found to express carboxypeptidase D and M, which reportedly to inactivate C5a. Also, the RCC cells stably transfected with Lenti-C5a produced larger transgrafted tumors in nude mice compared with the non-transfected or control virus transfected cells. In addition, over-expression of C5a significantly increased the expression and phosphorylation of STAT3 as well as the phosphorylated JNK level. Furthermore, the effect of C5a over-expression on RCC cells' proliferation, migration, and invasion could be blocked by Stattic, a STAT3-specific inhibitor. Conclusion Chronic over-activation of C5a/C5aR1 axis could directly increase RCC cells' proliferation, migration, and invasion and thus contribute directly to the progression of the disease. Over-activation of STAT3 pathway is among the underlying mechanism.
Collapse
Affiliation(s)
- Jing-Min Zheng
- Department of Urology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, People's Republic of China
| | - Han-Xi Zhou
- Department of Urology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, People's Republic of China
| | - Hong-Yuan Yu
- Department of Urology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, People's Republic of China
| | - Yu-Hui Xia
- Department of Pathology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, People's Republic of China
| | - Qing-Xin Yu
- Department of Pathology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, People's Republic of China
| | - Hang-Shuai Qu
- Department of Urology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, People's Republic of China
| | - Jia-Qian Bao
- Department of Urology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, People's Republic of China
| |
Collapse
|
16
|
Haffner-Luntzer M, Fischer V, Ignatius A. Differences in Fracture Healing Between Female and Male C57BL/6J Mice. Front Physiol 2021; 12:712494. [PMID: 34434120 PMCID: PMC8381649 DOI: 10.3389/fphys.2021.712494] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
Background Mice are increasingly used in fracture healing research because of the opportunity to use transgenic animals. While both, male and female mice are employed, there is no consensus in the literature whether fracture healing differs between both sexes. Therefore, the aim of the present study was to analyze diaphyseal fracture healing in female and male C57BL/6J mice, a commonly used mouse strain in bone research. Methods For that purpose, 12-week-old Female (17–20 g) and Male mice (22–26 g) received a standardized femur midshaft osteotomy stabilized by an external fixator. Mice were euthanized 10 and 21 days after fracture and bone healing was analyzed by biomechanical testing, μCT, histology, immunohistochemistry and qPCR. Results Ten days after fracture, Male mice displayed significantly more cartilage but less fibrous tissue in the fracture callus compared to Female mice, whereas the amount of bone did not differ. At day 21, Male mice showed a significantly larger fracture callus compared to Female mice. The relative amount of bone in the fracture callus did not significantly differ between both sexes, whereas its tissue mineral density was significantly higher in Male mice on day 21, indicating more mature bone and slightly more rapid fracture healing. These results were confirmed by a significantly greater absolute bending stiffness of the fractured femurs of Male mice on day 21. On the molecular level, Male mice displayed increased active β-catenin expression in the fracture callus, whereas estrogen receptor α (ERα) expression was lower. Conclusion These results suggest that Male mice display more rapid fracture healing with more prominent cartilaginous callus formation. This might be due to the higher weight of Male mice, resulting in increased mechanical loading of the fracture. Furthermore, Male mice displayed significantly greater activation of osteoanabolic Wnt/β-catenin signaling, which might also contribute to more rapid bone regeneration.
Collapse
Affiliation(s)
- Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University Medical Centre Ulm, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopaedic Research and Biomechanics, University Medical Centre Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Medical Centre Ulm, Ulm, Germany
| |
Collapse
|
17
|
Longoni A, Pennings I, Cuenca Lopera M, van Rijen MHP, Peperzak V, Rosenberg AJWP, Levato R, Gawlitta D. Endochondral Bone Regeneration by Non-autologous Mesenchymal Stem Cells. Front Bioeng Biotechnol 2020; 8:651. [PMID: 32733861 PMCID: PMC7363768 DOI: 10.3389/fbioe.2020.00651] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/27/2020] [Indexed: 12/31/2022] Open
Abstract
Mimicking endochondral bone formation is a promising strategy for bone regeneration. To become a successful therapy, the cell source is a crucial translational aspect. Typically, autologous cells are used. The use of non-autologous mesenchymal stromal cells (MSCs) represents an interesting alternative. Nevertheless, non-autologous, differentiated MSCs may trigger an undesired immune response, hampering bone regeneration. The aim of this study was to unravel the influence of the immune response on endochondral bone regeneration, when using xenogeneic (human) or allogeneic (Dark Agouti) MSCs. To this end, chondrogenically differentiated MSCs embedded in a collagen carrier were implanted in critical size femoral defects of immunocompetent Brown Norway rats. Control groups were included with syngeneic/autologous (Brown Norway) MSCs or a cell-free carrier. The amount of neo-bone formation was proportional to the degree of host-donor relatedness, as no full bridging of the defect was observed in the xenogeneic group whereas 2/8 and 7/7 bridges occurred in the allogeneic and the syngeneic group, respectively. One week post-implantation, the xenogeneic grafts were invaded by pro-inflammatory macrophages, T lymphocytes, which persisted after 12 weeks, and anti-human antibodies were developed. The immune response toward the allogeneic graft was comparable to the one evoked by the syngeneic implants, aside from an increased production of alloantibodies, which might be responsible for the more heterogeneous bone formation. Our results demonstrate for the first time the feasibility of using non-autologous MSC-derived chondrocytes to elicit endochondral bone regeneration in vivo. Nevertheless, the pronounced immune response and the limited bone formation observed in the xenogeneic group undermine the clinical relevance of this group. On the contrary, although further research on how to achieve robust bone formation with allogeneic cells is needed, they may represent an alternative to autologous transplantation.
Collapse
Affiliation(s)
- Alessia Longoni
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| | - I Pennings
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| | - Marta Cuenca Lopera
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - M H P van Rijen
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands.,Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Victor Peperzak
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - A J W P Rosenberg
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Riccardo Levato
- Regenerative Medicine Center Utrecht, Utrecht, Netherlands.,Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
18
|
Cai Y, Yang C, Yu X, Qian J, Dai M, Wang Y, Qin C, Lai W, Chen S, Wang T, Zhou J, Ma N, Zhang Y, Zhang R, Shen N, Xie X, Du C. Deficiency of β-Arrestin 2 in Dendritic Cells Contributes to Autoimmune Diseases. THE JOURNAL OF IMMUNOLOGY 2018; 202:407-420. [PMID: 30541881 DOI: 10.4049/jimmunol.1800261] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 11/13/2018] [Indexed: 12/30/2022]
Abstract
Altered migration and immune responses of dendritic cells (DCs) lead to inflammatory and autoimmune diseases. Our studies demonstrated that β-arrestin 2 deficiency promoted migration and cytokine production of mouse bone marrow-derived DCs. We further found that β-arrestin 2 directly interacted with Zbtb46, a DC-specific transcription factor. What's more, our results suggested that the interaction between β-arrestin 2 and Zbtb46 might negatively regulate DC migration. Using RNA sequencing, we indicated that genes CD74, NR4A1, and ZFP36 might be the target genes regulated by the interaction between β-arrestin 2 and Zbtb46. Mice with selective deficiency of β-arrestin 2 in DCs developed severer experimental autoimmune encephalomyelitis with more DC infiltration in the CNS and increased IL-6 in serum. In the systemic lupus erythematosus mice model, Arrb2fl/fl Itgax-cre+ mice were prone to exacerbation of lupus nephritis with a higher level of IL-6 and DC accumulation. Taken together, our study identified β-arrestin 2 as a new regulator of DC migration and immune properties, providing new insights into the mechanisms underlying the development of autoimmune disease.
Collapse
Affiliation(s)
- Yingying Cai
- Putuo District People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Cuixia Yang
- Putuo District People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiaohan Yu
- Department of Respiratory and Gastroenterology, Yingshan People's Hospital, Yingshan, Hubei 436700, China
| | - Jie Qian
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Min Dai
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Yan Wang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China; and
| | - Chaoyan Qin
- Putuo District People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Weiming Lai
- Putuo District People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Shuai Chen
- Putuo District People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Tingting Wang
- Putuo District People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jinfeng Zhou
- Putuo District People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Ningjia Ma
- Putuo District People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yue Zhang
- Putuo District People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Ru Zhang
- Putuo District People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Nan Shen
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Xin Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Changsheng Du
- Putuo District People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China;
| |
Collapse
|
19
|
Mödinger Y, Teixeira GQ, Neidlinger-Wilke C, Ignatius A. Role of the Complement System in the Response to Orthopedic Biomaterials. Int J Mol Sci 2018; 19:ijms19113367. [PMID: 30373272 PMCID: PMC6274916 DOI: 10.3390/ijms19113367] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/22/2022] Open
Abstract
Various synthetic biomaterials are used to replace lost or damaged bone tissue that, more or less successfully, osseointegrate into the bone environment. Almost all biomaterials used in orthopedic medicine activate the host-immune system to a certain degree. The complement system, which is a crucial arm of innate immunity, is rapidly activated by an implanted foreign material into the human body, and it is intensely studied regarding blood-contacting medical devices. In contrast, much less is known regarding the role of the complement system in response to implanted bone biomaterials. However, given the increasing knowledge of the complement regulation of bone homeostasis, regeneration, and inflammation, complement involvement in the immune response following biomaterial implantation into bone appears very likely. Moreover, bone cells can produce complement factors and are target cells of activated complement. Therefore, new bone formation or bone resorption around the implant area might be greatly influenced by the complement system. This review aims to summarize the current knowledge on biomaterial-mediated complement activation, with a focus on materials primarily used in orthopedic medicine. In addition, methods to modify the interactions between the complement system and bone biomaterials are discussed, which might favor osseointegration and improve the functionality of the device.
Collapse
Affiliation(s)
- Yvonne Mödinger
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| | - Graciosa Q Teixeira
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| | - Cornelia Neidlinger-Wilke
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Centre for Trauma Research Ulm (ZTF Ulm), University of Ulm, D-89081 Ulm, Germany.
| |
Collapse
|
20
|
Mödinger Y, Rapp AE, Vikman A, Ren Z, Fischer V, Bergdolt S, Haffner-Luntzer M, Song WC, Lambris JD, Huber-Lang M, Neidlinger-Wilke C, Brenner RE, Ignatius A. Reduced Terminal Complement Complex Formation in Mice Manifests in Low Bone Mass and Impaired Fracture Healing. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:147-161. [PMID: 30339839 DOI: 10.1016/j.ajpath.2018.09.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/01/2018] [Accepted: 09/17/2018] [Indexed: 12/20/2022]
Abstract
The terminal complement complex (TCC) is formed on activation of the complement system, a crucial arm of innate immunity. TCC formation on cell membranes results in a transmembrane pore leading to cell lysis. In addition, sublytic TCC concentrations can modulate various cellular functions. TCC-induced effects may play a role in the pathomechanisms of inflammatory disorders of the bone, including rheumatoid arthritis and osteoarthritis. In this study, we investigated the effect of the TCC on bone turnover and repair. Mice deficient for complement component 6 (C6), an essential component for TCC assembly, and mice with a knockout of CD59, which is a negative regulator of TCC formation, were used in this study. The bone phenotype was analyzed in vivo, and bone cell behavior was analyzed ex vivo. In addition, the mice were subjected to a femur osteotomy. Under homeostatic conditions, C6-deficient mice displayed a reduced bone mass, mainly because of increased osteoclast activity. After femur fracture, the inflammatory response was altered and bone formation was disturbed, which negatively affected the healing outcome. By contrast, CD59-knockout mice only displayed minor skeletal alterations and uneventful bone healing, although the early inflammatory reaction to femur fracture was marginally enhanced. These results demonstrate that TCC-mediated effects regulate bone turnover and promote an adequate response to fracture, contributing to an uneventful healing outcome.
Collapse
Affiliation(s)
- Yvonne Mödinger
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anna E Rapp
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anna Vikman
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Zhaozhou Ren
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Stephanie Bergdolt
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Wen-Chao Song
- Department of Pharmacology and Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University Medical Center, Ulm, Germany
| | | | - Rolf E Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Trauma Research Center Ulm, Ulm University Medical Center, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
21
|
Mödinger Y, Rapp A, Pazmandi J, Vikman A, Holzmann K, Haffner-Luntzer M, Huber-Lang M, Ignatius A. C5aR1 interacts with TLR2 in osteoblasts and stimulates the osteoclast-inducing chemokine CXCL10. J Cell Mol Med 2018; 22:6002-6014. [PMID: 30247799 PMCID: PMC6237570 DOI: 10.1111/jcmm.13873] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 07/13/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
The anaphylatoxin C5a is generated upon activation of the complement system, a crucial arm of innate immunity. C5a mediates proinflammatory actions via the C5a receptor C5aR1 and thereby promotes host defence, but also modulates tissue homeostasis. There is evidence that the C5a/C5aR1 axis is critically involved both in physiological bone turnover and in inflammatory conditions affecting bone, including osteoarthritis, periodontitis, and bone fractures. C5a induces the migration and secretion of proinflammatory cytokines of osteoblasts. However, the underlying mechanisms remain elusive. Therefore, in this study we aimed to determine C5a‐mediated downstream signalling in osteoblasts. Using a whole‐genome microarray approach, we demonstrate that C5a activates mitogen‐activated protein kinases (MAPKs) and regulates the expression of genes involved in pathways related to insulin, transforming growth factor‐β and the activator protein‐1 transcription factor. Interestingly, using coimmunoprecipitation, we found an interaction between C5aR1 and Toll‐like receptor 2 (TLR2) in osteoblasts. The C5aR1‐ and TLR2‐signalling pathways converge on the activation of p38 MAPK and the generation of C‐X‐C motif chemokine 10, which functions, among others, as an osteoclastogenic factor. In conclusion, C5a‐stimulated osteoblasts might modulate osteoclast activity and contribute to immunomodulation in inflammatory bone disorders.
Collapse
Affiliation(s)
- Yvonne Mödinger
- Institute of Orthopaedic Research and Biomechanics, Trauma Research Center Ulm, Ulm University Hospital, Ulm, Germany
| | - Anna Rapp
- Institute of Orthopaedic Research and Biomechanics, Trauma Research Center Ulm, Ulm University Hospital, Ulm, Germany
| | - Julia Pazmandi
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Anna Vikman
- Institute of Orthopaedic Research and Biomechanics, Trauma Research Center Ulm, Ulm University Hospital, Ulm, Germany
| | | | - Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, Trauma Research Center Ulm, Ulm University Hospital, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, Trauma Research Center Ulm, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
22
|
Wang D, Liu Y, Yang X, Zhou J. Hypoxic preconditioning enhances cell hypoxia tolerance and correlated lncRNA and mRNA analysis. Life Sci 2018; 208:46-54. [DOI: 10.1016/j.lfs.2018.07.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/23/2018] [Accepted: 07/07/2018] [Indexed: 01/04/2023]
|
23
|
Mödinger Y, Löffler B, Huber-Lang M, Ignatius A. Complement involvement in bone homeostasis and bone disorders. Semin Immunol 2018; 37:53-65. [DOI: 10.1016/j.smim.2018.01.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/17/2018] [Accepted: 01/22/2018] [Indexed: 12/12/2022]
|
24
|
Kaiser K, Prystaz K, Vikman A, Haffner-Luntzer M, Bergdolt S, Strauss G, Waetzig GH, Rose-John S, Ignatius A. Pharmacological inhibition of IL-6 trans-signaling improves compromised fracture healing after severe trauma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2018; 391:523-536. [PMID: 29497762 PMCID: PMC5889421 DOI: 10.1007/s00210-018-1483-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/19/2018] [Indexed: 01/01/2023]
Abstract
Patients with multiple injuries frequently suffer bone fractures and are at high risk to develop fracture healing complications. Because of its key role both in systemic posttraumatic inflammation and fracture healing, the pleiotropic cytokine interleukin-6 (IL-6) may be involved in the pathomechanisms of trauma-induced compromised fracture healing. IL-6 signals are transmitted by two different mechanisms: classic signaling via the membrane-bound receptor (mIL-6R) and trans-signaling via its soluble form (sIL-6R). Herein, we investigated whether IL-6 classic and trans-signaling play different roles in bone regeneration after severe injury. Twelve-week-old C57BL/6J mice underwent combined femur osteotomy and thoracic trauma. To study the function of IL-6, either an anti-IL-6 antibody, which inhibits both IL-6 classic and trans-signaling, or a soluble glycoprotein 130 fusion protein (sgp130Fc), which selectively blocks trans-signaling, were injected 30 min and 48 h after surgery. Bone healing was assessed using cytokine analyses, flow cytometry, histology, micro-computed tomography, and biomechanical testing. Selective inhibition of IL-6 trans-signaling significantly improved the fracture healing outcome after combined injury, as confirmed by accelerated cartilage-to-bone transformation, enhanced bony bridging of the fracture gap and improved mechanical callus properties. In contrast, global IL-6 inhibition did not affect compromised fracture healing. These data suggest that classic signaling may mediate beneficial effects on bone repair after severe injury. Selective inhibition of IL-6 trans-signaling might have therapeutic potential to treat fracture healing complications in patients with concomitant injuries.
Collapse
Affiliation(s)
- Kathrin Kaiser
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, University Medical Center Ulm, 89081, Ulm, Germany
| | - Katja Prystaz
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, University Medical Center Ulm, 89081, Ulm, Germany
| | - Anna Vikman
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, University Medical Center Ulm, 89081, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, University Medical Center Ulm, 89081, Ulm, Germany
| | - Stephanie Bergdolt
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, University Medical Center Ulm, 89081, Ulm, Germany
| | - Gudrun Strauss
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, 89075, Ulm, Germany
| | | | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University of Kiel, 24118, Kiel, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm, University Medical Center Ulm, 89081, Ulm, Germany.
| |
Collapse
|
25
|
Silawal S, Triebel J, Bertsch T, Schulze-Tanzil G. Osteoarthritis and the Complement Cascade. CLINICAL MEDICINE INSIGHTS. ARTHRITIS AND MUSCULOSKELETAL DISORDERS 2018; 11:1179544117751430. [PMID: 29434479 PMCID: PMC5805003 DOI: 10.1177/1179544117751430] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022]
Abstract
Accumulating evidence demonstrates that complement activation is involved in the pathogenesis of osteoarthritis (OA). However, the intimate complement regulation and cross talk with other signaling pathways in joint-associated tissues remain incompletely understood. Recent insights are summarized and discussed here, to put together a more comprehensive picture of complement involvement in OA pathogenesis. Complement is regulated by several catabolic and inflammatory mediators playing a key role in OA. It seems to be involved in many processes observed during OA development and progression, such as extracellular cartilage matrix (ECM) degradation, chondrocyte and synoviocyte inflammatory responses, cell lysis, synovitis, disbalanced bone remodeling, osteophyte formation, and stem cell recruitment, as well as cartilage angiogenesis. In reverse, complement can be activated by various ECM components and their cleavage products, which are released during OA-associated cartilage degradation. There are, however, some other cartilage ECM components that can inhibit complement, underlining the diverse effects of ECM on the complement activation. It is hypothesized that complement might also be directly activated by mechanical stress, thereby contributing to OA. The question arises whether keeping the complement activation in balance could represent a future therapeutic strategy in OA treatment and in the prevention of its progression.
Collapse
Affiliation(s)
- Sandeep Silawal
- Department of Anatomy, Paracelsus Medical University, Nuremberg, Germany
- Institute of Anatomy, Paracelsus Medical University, Salzburg, Germany
| | - Jakob Triebel
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital, Paracelsus Medical University, Nuremberg, Germany
| | - Thomas Bertsch
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital, Paracelsus Medical University, Nuremberg, Germany
| | - Gundula Schulze-Tanzil
- Department of Anatomy, Paracelsus Medical University, Nuremberg, Germany
- Institute of Anatomy, Paracelsus Medical University, Salzburg, Germany
| |
Collapse
|
26
|
Complement receptors C5aR1 and C5aR2 act differentially during the early immune response after bone fracture but are similarly involved in bone repair. Sci Rep 2017; 7:14061. [PMID: 29070810 PMCID: PMC5656620 DOI: 10.1038/s41598-017-14444-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/10/2017] [Indexed: 12/31/2022] Open
Abstract
Severely injured patients frequently suffer compromised fracture healing because of systemic post-traumatic inflammation. An important trigger of the posttraumatic immune response is the complement anaphylatoxin C5a, which acts via two receptors, C5aR1 and C5aR2, expressed on immune and bone cells. The blockade of C5a-mediated inflammation during the early inflammatory phase was demonstrated to improve fracture healing after severe injury. However, the distinct roles of the two complement receptors C5aR1 and C5aR2 in bone has to date not been studied. Here, we investigated bone turnover and regeneration in mice lacking either C5aR1 or C5aR2 in a model of isolated fracture and after severe injury, combining the fracture with an additional thoracic trauma. Both C5aR1−/− and C5aR2−/− mice displayed an increased bone mass compared to wild-type controls due to reduced osteoclast formation and increased osteoblast numbers, respectively. Following fracture, the inflammatory response was differently affected in these strains: It was decreased in C5aR1−/− mice but enhanced in C5aR2−/− mice. Both strains exhibited impaired fracture healing, disturbed osteoclastogenesis and delayed cartilage-to-bone transformation. Thus, our data suggest that C5aR1 and C5aR2 differentially regulate the immune response after fracture and are required for effective cartilage-to-bone transformation in the fracture callus and for undisturbed bone healing.
Collapse
|