11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      MyD88 and IL-1R signaling drive antibacterial immunity and osteoclast-driven bone loss during Staphylococcus aureus osteomyelitis

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Staphylococcus aureus is able to infect virtually all organ systems and is a frequently isolated etiologic agent of osteomyelitis, a common and debilitating invasive infection of bone. Treatment of osteomyelitis requires invasive surgical procedures and prolonged antibiotic therapy, yet is frequently unsuccessful due to extensive pathogen-induced bone damage that can limit antibiotic penetration and immune cell influx to the infectious focus. We previously established that S. aureus triggers profound alterations in bone remodeling in a murine model of osteomyelitis, in part through the production of osteolytic toxins. However, staphylococcal strains lacking osteolytic toxins still incite significant bone destruction, suggesting that host immune responses are also major drivers of pathologic bone remodeling during osteomyelitis. The objective of this study was to identify host immune pathways that contribute to antibacterial immunity during S. aureus osteomyelitis, and to define how these immune responses alter bone homeostasis and contribute to bone destruction. We specifically focused on the interleukin-1 receptor (IL-1R) and downstream adapter protein MyD88 given the prominent role of this signaling pathway in both antibacterial immunity and osteo-immunologic crosstalk. We discovered that while IL-1R signaling is necessary for local control of bacterial replication during osteomyelitis, it also contributes to bone loss during infection. Mechanistically, we demonstrate that S. aureus enhances osteoclastogenesis of myeloid precursors in vitro, and increases the abundance of osteoclasts residing on bone surfaces in vivo. This enhanced osteoclast abundance translates to trabecular bone loss, and is dependent on intact IL-1R signaling. Collectively, these data define IL-1R signaling as a critical component of the host response to S. aureus osteomyelitis, but also demonstrate that IL-1R-dependent immune responses trigger collateral bone damage through activation of osteoclast-mediated bone resorption.

          Author summary

          Osteomyelitis is a common, debilitating infection of bone that rarely resolves without prolonged antibiotics and invasive surgical procedures. This study explores the role of host inflammation during osteomyelitis. Our findings highlight innate immune responses that are critical for control of S. aureus burdens in bone, prevention of bacterial dissemination, and death. Conversely, these same immune pathways were found to contribute to disease pathogenesis by activating infection-associated bone loss. Such bone loss is associated with detrimental outcomes during osteomyelitis, including pathologic fractures and irreversible changes in bone growth plates. Alterations in bone remodeling that occur during S. aureus osteomyelitis are multifaceted, driven by bacterial toxins and inflammation-mediated changes. We found that inflammation incited by S. aureus in bone contributes to pathologic bone loss. We highlight a mechanism by which S. aureus stimulates formation of bone-resorbing osteoclasts both in vitro and in vivo. Our data illustrate connections between innate immune signaling pathways and bone homeostasis. These connections may be relevant to other autoinflammatory bone and joint conditions characterized by bone loss, such as rheumatoid arthritis. Collectively, this work outlines the fine balance between promotion of bacterial clearance and protection from collateral tissue damage.

          Related collections

          Most cited references74

          • Record: found
          • Abstract: found
          • Article: not found

          Osteomyelitis.

          Bone and joint infections are painful for patients and frustrating for both them and their doctors. The high success rates of antimicrobial therapy in most infectious diseases have not yet been achieved in bone and joint infections owing to the physiological and anatomical characteristics of bone. The key to successful management is early diagnosis, including bone sampling for microbiological and pathological examination to allow targeted and long-lasting antimicrobial therapy. The various types of osteomyelitis require differing medical and surgical therapeutic strategies. These types include, in order of decreasing frequency: osteomyelitis secondary to a contiguous focus of infection (after trauma, surgery, or insertion of a joint prosthesis); that secondary to vascular insufficiency (in diabetic foot infections); or that of haematogenous origin. Chronic osteomyelitis is associated with avascular necrosis of bone and formation of sequestrum (dead bone), and surgical debridement is necessary for cure in addition to antibiotic therapy. By contrast, acute osteomyelitis can respond to antibiotics alone. Generally, a multidisciplinary approach is required for success, involving expertise in orthopaedic surgery, infectious diseases, and plastic surgery, as well as vascular surgery, particularly for complex cases with soft-tissue loss.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL.

            Osteoclasts, the multinucleated cells that resorb bone, develop from hematopoietic cells of monocyte/macrophage lineage. Osteoclast-like cells (OCLs) are formed by coculturing spleen cells with osteoblasts or bone marrow stromal cells in the presence of bone-resorbing factors. The cell-to-cell interaction between osteoblasts/stromal cells and osteoclast progenitors is essential for OCL formation. Recently, we purified and molecularly cloned osteoclastogenesis-inhibitory factor (OCIF), which was identical to osteoprotegerin (OPG). OPG/OCIF is a secreted member of the tumor necrosis factor receptor family and inhibits osteoclastogenesis by interrupting the cell-to-cell interaction. Here we report the expression cloning of a ligand for OPG/OCIF from a complementary DNA library of mouse stromal cells. The protein was found to be a member of the membrane-associated tumor necrosis factor ligand family and induced OCL formation from osteoclast progenitors. A genetically engineered soluble form containing the extracellular domain of the protein induced OCL formation from spleen cells in the absence of osteoblasts/stromal cells. OPG/OCIF abolished the OCL formation induced by the protein. Expression of its gene in osteoblasts/stromal cells was up-regulated by bone-resorbing factors. We conclude that the membrane-bound protein is osteoclast differentiation factor (ODF), a long-sought ligand mediating an essential signal to osteoclast progenitors for their differentiation into osteoclasts. ODF was found to be identical to TRANCE/RANKL, which enhances T-cell growth and dendritic-cell function. ODF seems to be an important regulator in not only osteoclastogenesis but also immune system.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Gut microbiota induce IGF-1 and promote bone formation and growth.

              Appreciation of the role of the gut microbiome in regulating vertebrate metabolism has exploded recently. However, the effects of gut microbiota on skeletal growth and homeostasis have only recently begun to be explored. Here, we report that colonization of sexually mature germ-free (GF) mice with conventional specific pathogen-free (SPF) gut microbiota increases both bone formation and resorption, with the net effect of colonization varying with the duration of colonization. Although colonization of adult mice acutely reduces bone mass, in long-term colonized mice, an increase in bone formation and growth plate activity predominates, resulting in equalization of bone mass and increased longitudinal and radial bone growth. Serum levels of insulin-like growth factor 1 (IGF-1), a hormone with known actions on skeletal growth, are substantially increased in response to microbial colonization, with significant increases in liver and adipose tissue IGF-1 production. Antibiotic treatment of conventional mice, in contrast, decreases serum IGF-1 and inhibits bone formation. Supplementation of antibiotic-treated mice with short-chain fatty acids (SCFAs), products of microbial metabolism, restores IGF-1 and bone mass to levels seen in nonantibiotic-treated mice. Thus, SCFA production may be one mechanism by which microbiota increase serum IGF-1. Our study demonstrates that gut microbiota provide a net anabolic stimulus to the skeleton, which is likely mediated by IGF-1. Manipulation of the microbiome or its metabolites may afford opportunities to optimize bone health and growth.
                Bookmark

                Author and article information

                Contributors
                Role: ConceptualizationRole: Formal analysisRole: Funding acquisitionRole: InvestigationRole: MethodologyRole: ValidationRole: VisualizationRole: Writing – original draftRole: Writing – review & editing
                Role: Formal analysisRole: InvestigationRole: ValidationRole: Writing – review & editing
                Role: InvestigationRole: Writing – review & editing
                Role: InvestigationRole: Writing – review & editing
                Role: InvestigationRole: ValidationRole: Writing – review & editing
                Role: Investigation
                Role: ConceptualizationRole: Funding acquisitionRole: InvestigationRole: MethodologyRole: Project administrationRole: ResourcesRole: SupervisionRole: Writing – original draftRole: Writing – review & editing
                Role: Editor
                Journal
                PLoS Pathog
                PLoS Pathog
                plos
                plospath
                PLoS Pathogens
                Public Library of Science (San Francisco, CA USA )
                1553-7366
                1553-7374
                12 April 2019
                April 2019
                : 15
                : 4
                : e1007744
                Affiliations
                [1 ] Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
                [2 ] Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
                [3 ] Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
                [4 ] Vanderbilt Institute for Infection, Immunology and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
                [5 ] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
                Johns Hopkins School of Medicine, UNITED STATES
                Author notes

                The authors have declared that no competing interests exist.

                [¤]

                Current address: College of Medicine and Life Sciences, The University of Toledo, Toledo, Ohio, United States of America

                Author information
                http://orcid.org/0000-0002-7975-0503
                http://orcid.org/0000-0002-9242-0461
                http://orcid.org/0000-0002-3710-8037
                http://orcid.org/0000-0001-9969-6607
                Article
                PPATHOGENS-D-18-02431
                10.1371/journal.ppat.1007744
                6481883
                30978245
                a5e09b86-3ee9-4e35-b459-3e87872daaf3
                © 2019 Putnam et al

                This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                : 19 December 2018
                : 1 April 2019
                Page count
                Figures: 8, Tables: 0, Pages: 31
                Funding
                JEC is supported by National Institute of Allergy and Infectious Diseases ( https://www.niaid.nih.gov) grants 1R01AI132560 and 5K08AI113107 and a Career Award for Medical Scientists from the Burroughs Wellcome Fund ( https://www.bwfund.org). NEP is supported by the National Institute of Allergy and Infectious Diseases ( https://www.niaid.nih.gov) grant 1F31AI133926. CAF was supported through the National Institute of General Medical Sciences ( https://www.nigms.nih.gov) Medical Scientist Training Program grant T32GM007347 and is currently supported by the National Institute of Allergy and Infectious Diseases ( https://www.niaid.nih.gov) grant 1F30AI138424. JRP is supported by the National Institute of Arthritis and Musculoskeletal and Skin Diseases ( https://www.niams.nih.gov) Interdisciplinary Training in Rheumatic Diseases grant 5T32AR059039. The research support for the μCT50 and computer cluster is maintained by National Institutes of Health ( https://www.nih.gov) grant S10RR027631. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article
                Custom metadata
                vor-update-to-uncorrected-proof
                2019-04-24
                All relevant data are within the manuscript and its Supporting Information files.

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article