1
|
Cao Y, Chen F, Zhu S, Zhu D, Qi H. Staphylococcus aureus infection initiates hypoxia-mediated STIP1 homology and U-box containing protein 1 upregulation to trigger osteomyelitis. Toxicon 2024; 248:108049. [PMID: 39059559 DOI: 10.1016/j.toxicon.2024.108049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Although little is known about the regulatory mechanisms underlying the pathogenesis of osteomyelitis caused by Staphylococcus aureus (S. aureus), hypoxia-inducible factor-1α (HIF-1α) and STIP1 homology and U-box containing protein 1 (STUB1) have been found to be up-regulated in both S. aureus infected MC3T3-E1 cells and in patients with osteomyelitis. HIF-1α directly targets STUB1 to induce its expression. In MC3T3-E1 cells infected with S. aureus, silencing HIF-1α and STUB1 and administering the hypoxia inhibitor IDF-11774 consistently increased the expression of OSX and RUNX2, as well as the levels of alizarin Red S and alkaline phosphatase activity. In a mouse model of osteomyelitis, S. aureus infection elevated HIF-1α expression and serum STUB1 levels. Interleukin (IL)-6, IL-1β, and C-reactive protein levels in serum were reduced after treatment with the hypoxia inhibitor IDF-11774. Following an infection with S. aureus, hypoxia was activated to cause STUB1 overexpression by directly targeting HIF-1α, ultimately causing osteomyelitis symptoms such as osteogenesis and mineralization defected and increased inflammation. This study presents a novel signaling cascade in the pathogenesis of osteomyelitis involving hypoxia/HIF-1α/STUB1. This signaling cascade may be a target for therapeutic interventions.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Pediatric Surgery, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Feng Chen
- Department of Pediatric, Luodian Hospital, Shanghai, China
| | - Suyue Zhu
- Department of Pediatric, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, China
| | - Dongsheng Zhu
- Department of Pediatric Surgery, The First People's Hospital of Lianyungang, Lianyungang, China.
| | - Han Qi
- Department of Surgery, The Second People's Hospital of Lianyungang, Lianyungang, China.
| |
Collapse
|
2
|
Lin Y, Yang M, Cheng C, Wu J, Yu B, Zhang X. Age-related dysregulation of CXCL9/10 in monocytes is linked to impaired innate immune responses in a mouse model of Staphylococcus aureus osteomyelitis. Cell Mol Life Sci 2024; 81:300. [PMID: 39001897 PMCID: PMC11335224 DOI: 10.1007/s00018-024-05311-2] [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: 12/27/2023] [Revised: 05/12/2024] [Accepted: 06/10/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Age-associated impairments in innate immunity are believed to be a causative factor responsible for severe pathogenesis of Staphylococcus aureus (S. aureus) infection in the bone tissue. However, the basis for age-associated decline in innate immune response upon S. aureus infection remains poorly understood. RESULTS Our transcriptional data (GEO: GSE166522) from a mouse model of S. aureus osteomyelitis show up-regulated CXCL9 and CXCL10 (CXCL9/10), which is further confirmed in vitro and in vivo by the present study. Notably, monocytes are a main source for CXCL9/10 production in bone marrow upon S. aureus challenge, but this response declines in middle-aged mice. Interestingly, conditional medium of bone marrow monocytes from middle-aged mice has a strikingly decreased effect on bactericidal functions of neutrophils and macrophages compares with that from young mice. We further show that activation of CXCL9/10-CXCR3 axis between monocytes and macrophages/neutrophils promotes the bactericidal function of the cells, whereas blocking the axis impairs such function. Importantly, treatment with either exogenous CXCL9 or CXCL10 in a middle-aged mice model enhances, while pharmacological inhibition of CXCR3 in young mice model impairs, bacterial clearance and bone marrow structure. CONCLUSIONS These findings demonstrate that bone marrow monocytes act as a critical promotor of innate immune response via the CXLCL9/10-CXCR3 axis upon S. aureus infection, and that the increased susceptibility to S. aureus infection in skeleton in an aged host may be largely attributable to the declined induction of CXCR9/10 in monocytes.
Collapse
Affiliation(s)
- Yihuang Lin
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No. 1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
- Department of Orthopaedics, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, China
| | - Mankai Yang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No. 1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Chubin Cheng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No. 1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Jichang Wu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No. 1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No. 1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Xianrong Zhang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No. 1838 North of Guangzhou Avenue, Guangzhou, Guangdong Province, 510515, China.
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China.
| |
Collapse
|
3
|
Volk CF, Proctor RA, Rose WE. The Complex Intracellular Lifecycle of Staphylococcus aureus Contributes to Reduced Antibiotic Efficacy and Persistent Bacteremia. Int J Mol Sci 2024; 25:6486. [PMID: 38928191 PMCID: PMC11203666 DOI: 10.3390/ijms25126486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Staphylococcus aureus bacteremia continues to be associated with significant morbidity and mortality, despite improvements in diagnostics and management. Persistent infections pose a major challenge to clinicians and have been consistently shown to increase the risk of mortality and other infectious complications. S. aureus, while typically not considered an intracellular pathogen, has been proven to utilize an intracellular niche, through several phenotypes including small colony variants, as a means for survival that has been linked to chronic, persistent, and recurrent infections. This intracellular persistence allows for protection from the host immune system and leads to reduced antibiotic efficacy through a variety of mechanisms. These include antimicrobial resistance, tolerance, and/or persistence in S. aureus that contribute to persistent bacteremia. This review will discuss the challenges associated with treating these complicated infections and the various methods that S. aureus uses to persist within the intracellular space.
Collapse
Affiliation(s)
- Cecilia F. Volk
- Pharmacy Practice and Translational Research Division, School of Pharmacy, Pharmacy University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Richard A. Proctor
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Warren E. Rose
- Pharmacy Practice and Translational Research Division, School of Pharmacy, Pharmacy University of Wisconsin-Madison, Madison, WI 53705, USA;
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
4
|
Zhu D, Chen F, Qiang H, Qi H. SPA inhibits hBMSC osteogenic differentiation and M1 macrophage polarization by suppressing SETD2 in acute suppurative osteomyelitis. Sci Rep 2024; 14:12728. [PMID: 38830934 PMCID: PMC11148074 DOI: 10.1038/s41598-024-63219-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
To clarify the impact of SETD2 on macrophage function in pediatric patients with acute suppurative osteomyelitis and to elucidate the precise underlying mechanism. To gain insights into the potential functions of SETD2, a comprehensive study was conducted utilizing a co-culture model of human bone mesenchymal stem cells (hBMSCs) and bone marrow-derived macrophages (THP-1). A range of techniques were employed, including quantitative polymerase chain reaction, western blotting, ELISA, alkaline phosphatase activity assays, alizarin red S staining, luciferase reporter gene assays, and chromatin immunoprecipitation, to unravel the intricate interactions and molecular mechanisms involving SETD2 in this system. It was observed that SETD2 expression was reduced in THP-1 cells stimulated by staphylococcal protein A (SPA). Furthermore, the downregulation of SETD2 resulted in elevated M1 macrophage polarization and glycolysis, effects that were mitigated by SPA stimulation. Notably, SPA-stimulated THP-1 cells exhibited an increase in HIF-1α expression, which exhibited an inverse correlation with SETD2 levels. Moreover, it was discovered that SETD2 functioned as a catalyst for H3K36me3 and bound to the HIF-1α gene, which, in turn, regulated HIF-1α expression. Furthermore, the suppression of HIF-1α abrogated the consequences of SETD2 downregulation on glycolysis and M1 macrophage polarization. Lastly, the study demonstrated that M1 macrophage polarization serves as a mediator for BMP4's inhibitory effect on osteogenic differentiation of hBMSCs. This research has uncovered a previously unknown role of SETD2 in macrophages during osteomyelitis, revealing its significance in the pathogenesis of this condition. These findings suggest SETD2 as a novel target for the treatment of osteomyelitis.
Collapse
Affiliation(s)
- Dongsheng Zhu
- Department of Pediatric Surgery, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China.
| | - Feng Chen
- Department of Pediatric, Luodian Hospital, Shanghai, China
| | - Hongjia Qiang
- Department of Pediatric Surgery, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China.
| | - Han Qi
- Department of Emergency Surgery, The Second People's Hospital of , Lianyungang, Jiangsu Province, China.
| |
Collapse
|
5
|
Crosby HA, Keim K, Kwiecinski JM, Langouët-Astrié CJ, Oshima K, LaRivière WB, Schmidt EP, Horswill AR. Host-derived protease promotes aggregation of Staphylococcus aureus by cleaving the surface protein SasG. mBio 2024; 15:e0348323. [PMID: 38511930 PMCID: PMC11005337 DOI: 10.1128/mbio.03483-23] [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: 01/11/2024] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
Staphylococcus aureus is one of the leading causes of hospital-acquired infections, many of which begin following attachment and accumulation on indwelling medical devices or diseased tissue. These infections are often linked to the establishment of biofilms, but another often overlooked key characteristic allowing S. aureus to establish persistent infection is the formation of planktonic aggregates. Such aggregates are physiologically similar to biofilms and protect pathogens from innate immune clearance and increase antibiotic tolerance. The cell-wall-associated protein SasG has been implicated in biofilm formation via mechanisms of intercellular aggregation but the mechanism in the context of disease is largely unknown. We have previously shown that the expression of cell-wall-anchored proteins involved in biofilm formation is controlled by the ArlRS-MgrA regulatory cascade. In this work, we demonstrate that the ArlRS two-component system controls aggregation, by repressing the expression of sasG by activation of the global regulator MgrA. We also demonstrate that SasG must be proteolytically processed by a non-staphylococcal protease to induce aggregation and that strains expressing functional full-length sasG aggregate significantly upon proteolysis by a mucosal-derived host protease found in human saliva. We used fractionation and N-terminal sequencing to demonstrate that human trypsin within saliva cleaves within the A domain of SasG to expose the B domain and induce aggregation. Finally, we demonstrated that SasG is involved in virulence during mouse lung infection. Together, our data point to SasG, its processing by host proteases, and SasG-driven aggregation as important elements of S. aureus adaptation to the host environment.IMPORTANCEHere, we demonstrate that the Staphylococcus aureus surface protein SasG is important for cell-cell aggregation in the presence of host proteases. We show that the ArlRS two-component regulatory system controls SasG levels through the cytoplasmic regulator MgrA. We identified human trypsin as the dominant protease triggering SasG-dependent aggregation and demonstrated that SasG is important for S. aureus lung infection. The discovery that host proteases can induce S. aureus aggregation contributes to our understanding of how this pathogen establishes persistent infections. The observations in this study demonstrate the need to strengthen our knowledge of S. aureus surface adhesin function and processing, regulation of adhesin expression, and the mechanisms that promote biofilm formation to develop strategies for preventing chronic infections.
Collapse
Affiliation(s)
- Heidi A. Crosby
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Klara Keim
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jakub M. Kwiecinski
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Christophe J. Langouët-Astrié
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kaori Oshima
- Division of Pulmonary Sciences and Critical Care, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Wells B. LaRivière
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Eric P. Schmidt
- Division of Pulmonary Sciences and Critical Care, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado, USA
| |
Collapse
|
6
|
Hofstee MI, Siverino C, Saito M, Meghwani H, Tapia-Dean J, Arveladze S, Hildebrand M, Rangel-Moreno J, Riool M, Zeiter S, Zaat SAJ, Moriarty TF, Muthukrishnan G. Staphylococcus aureus Panton-Valentine Leukocidin worsens acute implant-associated osteomyelitis in humanized BRGSF mice. JBMR Plus 2024; 8:ziad005. [PMID: 38505530 PMCID: PMC10945728 DOI: 10.1093/jbmrpl/ziad005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/21/2024] Open
Abstract
Staphylococcus aureus is the most common pathogen that causes implant-associated osteomyelitis, a clinically incurable disease. Immune evasion of S. aureus relies on various mechanisms to survive within the bone niche, including the secretion of leukotoxins such as Panton-Valentine leukocidin (PVL). PVL is a pore-forming toxin exhibiting selective human tropism for C5a receptors (C5aR1 and C5aR2) and CD45 on neutrophils, monocytes, and macrophages. PVL is an important virulence determinant in lung, skin and soft tissue infections. The involvement of PVL in S. aureus pathogenesis during bone infections has not been studied extensively yet. To investigate this, humanized BALB/c Rag2-/-Il2rg-/-SirpaNODFlk2-/- (huBRGSF) mice were subjected to transtibial implant-associated osteomyelitis with community-acquired methicillin-resistant S. aureus (CA-MRSA) USA300 wild type strain (WT), an isogenic mutant lacking lukF/S-PV (Δpvl), or complemented mutant (Δpvl+pvl). Three days post-surgery, Δpvl-infected huBRGSF mice had a less severe infection compared to WT-infected animals as characterized by 1) improved clinical outcomes, 2) lower ex vivo bacterial bone burden, 3) absence of staphylococcal abscess communities (SACs) in their bone marrow, and 4) compromised MRSA dissemination to internal organs (liver, kidney, spleen, heart). Interestingly, Δpvl-infected huBRGSF mice had fewer human myeloid cells, neutrophils, and HLA-DR+ monocytes in the bone niche compared to WT-infected animals. Expectedly, a smaller fraction of human myeloid cells were apoptotic in the Δpvl-infected huBRGSF animals. Taken together, our study highlights the pivotal role of PVL during acute implant-associated osteomyelitis in humanized mice.
Collapse
Affiliation(s)
- Marloes I Hofstee
- AO Research Institute Davos, 7270 Davos, Switzerland
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam institute for Infection and Immunity, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | | | - Motoo Saito
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, United States
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14618, United States
| | - Himanshu Meghwani
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, United States
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14618, United States
| | | | | | | | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14620, United States
| | - Martijn Riool
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam institute for Infection and Immunity, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Trauma Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | | | - Sebastian A J Zaat
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam institute for Infection and Immunity, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | | | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, United States
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14618, United States
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, United States
| |
Collapse
|
7
|
Mandal S, Tannert A, Löffler B, Neugebauer U, Silva LB. Findaureus: An open-source application for locating Staphylococcus aureus in fluorescence-labelled infected bone tissue slices. PLoS One 2024; 19:e0296854. [PMID: 38295056 PMCID: PMC10830009 DOI: 10.1371/journal.pone.0296854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 12/20/2023] [Indexed: 02/02/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is a facultative pathogenic bacterium that can cause infections in various tissue types in humans. Fluorescence imaging techniques have been employed to visualize the bacteria in ex-vivo samples mostly in research, aiding in the understanding of the etiology of the pathogen. However, the multispectral images generated from fluorescence microscopes are complex, making it difficult to locate bacteria across image files, especially in consecutive planes with different imaging depths. To address this issue, we present Findaureus, an open-source application specifically designed to locate and extract critical information about bacteria, especially S. aureus. Due to the limited availability of datasets, we tested the application on a dataset comprising fluorescence-labelled infected mouse bone tissue images, achieving an accuracy of 95%. We compared Findaureus with other state-of-the-art image analysis tools and found that it performed better, given its specificity toward bacteria localization. The proposed approach has the potential to aid in medical research of bone infections and can be extended to other tissue and bacteria types in the future.
Collapse
Affiliation(s)
- Shibarjun Mandal
- Leibniz-Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), Jena, Germany
| | - Astrid Tannert
- Leibniz-Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Bettina Löffler
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Ute Neugebauer
- Leibniz-Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | | |
Collapse
|
8
|
Zhao Q, Yang N, Gu X, Li Y, Teng D, Hao Y, Lu H, Mao R, Wang J. High-Yield Preparation of American Oyster Defensin (AOD) via a Small and Acidic Fusion Tag and Its Functional Characterization. Mar Drugs 2023; 22:8. [PMID: 38276646 PMCID: PMC10821286 DOI: 10.3390/md22010008] [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: 11/04/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
The marine peptide, American oyster defensin (AOD), is derived from Crassostrea virginica and exhibits a potent bactericidal effect. However, recombinant preparation has not been achieved due to the high charge and hydrophobicity. Although the traditional fusion tags such as Trx and SUMO shield the effects of target peptides on the host, their large molecular weight (12-20 kDa) leads to the yields lower than 20% of the fusion protein. In this study, a short and acidic fusion tag was employed with a compact structure of only 1 kDa. Following 72 h of induction in a 5 L fermenter, the supernatant exhibited a total protein concentration of 587 mg/L. The recombinant AOD was subsequently purified through affinity chromatography and enterokinase cleavage, resulting in the final yield of 216 mg/L and a purity exceeding 93%. The minimum inhibitory concentrations (MICs) of AOD against Staphylococcus aureus, Staphylococcus epidermidis, and Streptococcus galactis ranged from 4 to 8 μg/mL. Moreover, time-killing curves indicated that AOD achieved a bactericidal rate of 99.9% against the clinical strain S. epidermidis G-81 within 0.5 h at concentrations of 2× and 4× MIC. Additionally, the activity of AOD was unchanged after treatment with artificial gastric fluid and intestinal fluid for 4 h. Biocompatibility testing demonstrated that AOD, at a concentration of 128 μg/mL, exhibited a hemolysis rate of less than 0.5% and a cell survival rate of over 83%. Furthermore, AOD's in vivo therapeutic efficacy against mouse subcutaneous abscess revealed its capability to restrain bacterial proliferation and reduce bacterial load, surpassing that of antibiotic lincomycin. These findings indicate AOD's potential for clinical usage.
Collapse
Affiliation(s)
- Qingyi Zhao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Na Yang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xinxi Gu
- Enzyme Engineering Laboratory, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Yuanyuan Li
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Da Teng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ya Hao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Haiqiang Lu
- Enzyme Engineering Laboratory, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Ruoyu Mao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Jianhua Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| |
Collapse
|
9
|
Huang S, Wen J, Zhang Y, Bai X, Cui ZK. Choosing the right animal model for osteomyelitis research: Considerations and challenges. J Orthop Translat 2023; 43:47-65. [PMID: 38094261 PMCID: PMC10716383 DOI: 10.1016/j.jot.2023.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 03/22/2024] Open
Abstract
Osteomyelitis is a debilitating bone disorder characterized by an inflammatory process involving the bone marrow, bone cortex, periosteum, and surrounding soft tissue, which can ultimately result in bone destruction. The etiology of osteomyelitis can be infectious, caused by various microorganisms, or noninfectious, such as chronic nonbacterial osteomyelitis (CNO) and chronic recurrent multifocal osteomyelitis (CRMO). Researchers have turned to animal models to study the pathophysiology of osteomyelitis. However, selecting an appropriate animal model that accurately recapitulates the human pathology of osteomyelitis while controlling for multiple variables that influence different clinical presentations remains a significant challenge. In this review, we present an overview of various animal models used in osteomyelitis research, including rodent, rabbit, avian/chicken, porcine, minipig, canine, sheep, and goat models. We discuss the characteristics of each animal model and the corresponding clinical scenarios that can provide a basic rationale for experimental selection. This review highlights the importance of selecting an appropriate animal model for osteomyelitis research to improve the accuracy of the results and facilitate the development of novel treatment and management strategies.
Collapse
Affiliation(s)
| | | | - Yiqing Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhong-Kai Cui
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
10
|
Hinz N, Butscheidt S, Jandl NM, Rohde H, Keller J, Beil FT, Hubert J, Rolvien T. Increased local bone turnover in patients with chronic periprosthetic joint infection. Bone Joint Res 2023; 12:644-653. [PMID: 37813394 PMCID: PMC10562080 DOI: 10.1302/2046-3758.1210.bjr-2023-0071.r1] [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] [Indexed: 10/11/2023] Open
Abstract
Aims The management of periprosthetic joint infection (PJI) remains a major challenge in orthopaedic surgery. In this study, we aimed to characterize the local bone microstructure and metabolism in a clinical cohort of patients with chronic PJI. Methods Periprosthetic femoral trabecular bone specimens were obtained from patients suffering from chronic PJI of the hip and knee (n = 20). Microbiological analysis was performed on preoperative joint aspirates and tissue specimens obtained during revision surgery. Microstructural and cellular bone parameters were analyzed in bone specimens by histomorphometry on undecalcified sections complemented by tartrate-resistant acid phosphatase immunohistochemistry. Data were compared with control specimens obtained during primary arthroplasty (n = 20) and aseptic revision (n = 20). Results PJI specimens exhibited a higher bone volume, thickened trabeculae, and increased osteoid parameters compared to both control groups, suggesting an accelerated bone turnover with sclerotic microstructure. On the cellular level, osteoblast and osteoclast parameters were markedly increased in the PJI cohort. Furthermore, a positive association between serum (CRP) but not synovial (white blood cell (WBC) count) inflammatory markers and osteoclast indices could be detected. Comparison between different pathogens revealed increased osteoclastic bone resorption parameters without a concomitant increase in osteoblasts in bone specimens from patients with Staphylococcus aureus infection, compared to those with detection of Staphylococcus epidermidis and Cutibacterium spp. Conclusion This study provides insights into the local bone metabolism in chronic PJI, demonstrating osteosclerosis with high bone turnover. The fact that Staphylococcus aureus was associated with distinctly increased osteoclast indices strongly suggests early surgical treatment to prevent periprosthetic bone alterations.
Collapse
Affiliation(s)
- Nico Hinz
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Trauma, Surgery and Sports Traumatology, BG Trauma Hospital Hamburg, Hamburg, Germany
| | - Sebastian Butscheidt
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nico M. Jandl
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Holger Rohde
- Instiute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Keller
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank T. Beil
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Hubert
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Rolvien
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
11
|
Mariano LC, Grenho L, Fernandes MH, de Sousa Gomes P. Integrative tissue, cellular and molecular responsiveness of an innovative ex vivo model of the Staphylococcus aureus-mediated bone infection. FASEB J 2023; 37:e23166. [PMID: 37650876 DOI: 10.1096/fj.202300287rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/21/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
Osteomyelitis is a pathological condition of the bone, frequently associated with the presence of infectious agents - namely Staphylococcus aureus - that induce inflammation and tissue destruction. Recent advances in the understanding of its pathophysiology and the identification of innovative therapeutic approaches were gathered from experimental in vitro and in vivo systems. However, cell culture models offer limited representativeness of the cellular functionality and the cell-cell and cell-matrix interactions, further failing to mimic the three-dimensional tissue organization; and animal models allow for limited mechanistic assessment given the complex nature of systemic and paracrine regulatory systems and are endorsed with ethical constraints. Accordingly, this study aims at the establishment and assessment of a new ex vivo bone infection model, upon the organotypic culture of embryonic chicken femurs colonized with S. aureus, highlighting the model responsiveness at the molecular, cellular, and tissue levels. Upon infection with distinct bacterial inoculums, data reported an initial exponential bacterial growth, followed by diminished metabolic activity. At the tissue level, evidence of S. aureus-mediated tissue destruction was attained and demonstrated through distinct methodologies, conjoined with decreased osteoblastic/osteogenic and increased osteoclastic/osteoclastogenic functionalities-representative of the osteomyelitis clinical course. Overall, the establishment and characterization of an innovative bone tissue infection model that is simple, reproducible, easily manipulated, cost-effective, and simulates many features of human osteomyelitis, further allowing the maintenance of the bone tissue's three-dimensional morphology and cellular arrangement, was achieved. Model responsiveness was further demonstrated, showcasing the capability to improve the research pipeline in bone tissue infection-related research.
Collapse
Affiliation(s)
- Lorena Castro Mariano
- BoneLab-Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, Porto, Portugal
- LAQV/REQUIMTE, University of Porto, Porto, Portugal
| | - Liliana Grenho
- BoneLab-Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, Porto, Portugal
- LAQV/REQUIMTE, University of Porto, Porto, Portugal
| | - Maria Helena Fernandes
- BoneLab-Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, Porto, Portugal
- LAQV/REQUIMTE, University of Porto, Porto, Portugal
| | - Pedro de Sousa Gomes
- BoneLab-Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, University of Porto, Porto, Portugal
- LAQV/REQUIMTE, University of Porto, Porto, Portugal
| |
Collapse
|
12
|
Gao L, Tang Z, Li T, Wang J. Myricetin exerts anti-biofilm activity and attenuates osteomyelitis by inhibiting the TLR2/MAPK pathway in experimental mice. Microb Pathog 2023; 182:106165. [PMID: 37224983 DOI: 10.1016/j.micpath.2023.106165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/13/2023] [Accepted: 05/18/2023] [Indexed: 05/26/2023]
Abstract
AIMS To evaluate the potential of Myricetin against S.aureus induced osteomyelitis. BACKGROUND Osteomyelitis is infected condition of bone by micro-organisms. The mitogen-activated protein kinase (MAPK), inflammatory cytokines and Toll-like receptor-2 (TLR-2) pathway are mainly involved in osteomyelitis. Myricetin is a plant-food derived flavonoid which shows anti-inflammatory activity. OBJECTIVE In the present study, we evaluated the potential of Myricetin against S.aureus induced osteomyelitis. MC3T3-E1 cells were used for in vitro studies. METHOD Murine model of osteomyelitis was developed in BALB/c mice by injecting S.aureus in the medullary cavity of the femur. The mice were studied for bone destruction, anti-biofilm activity, osteoblast growth markers alkaline phosphatase (ALP), osteopontin (OCN) and collagen type-I (COLL-1) were studied by RT-PCR, ELISA analysis for levels of proinflammatory factors CRP, IL-6 and IL-1β. Expression of proteins by Western blot analysis and anti-biofilm effect by Sytox green dye fluorescence assay. Target confirmation was done by performing in silico docking analysis. RESULTS Myricetin reduced bone destruction in osteomyelitis induced mice. The treatment decreased bone levels of ALP, OCN, COLL-1 and TLR2. Myricetin decreased serum levels of CRP, IL-6 and IL-1β. The treatment suppressed activation of MAPK pathway and showed anti-biofilm effect. Docking studies suggested high binding affinity of Myricetin with MAPK protein in silico, by showing lower binding energies. CONCLUSION Myricetin suppresses osteomyelitis by inhibiting ALP, OCN, COLL-1 via the TLR2 and MAPK pathway involving inhibition of biofilm formation. In silico studies suggested MAPK as potential binding protein for myricetin.
Collapse
Affiliation(s)
- Lei Gao
- Department of Orthopaedic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China.
| | - Zhiping Tang
- Clinical Lab, He Bei General Hospital, Shi Jia Zhuang, 050051, China.
| | - Tianbo Li
- Department of Orthopaedic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China.
| | - Jiangning Wang
- Department of Orthopaedic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China.
| |
Collapse
|
13
|
Aguilera-Correa JJ, Salinas B, González-Arjona M, de Pablo D, Muñoz P, Bouza E, Fernández Aceñero MJ, Esteban J, Desco M, Cussó L. Positron Emission Tomography-Computed Tomography and Magnetic Resonance Imaging Assessments in a Mouse Model of Implant-Related Bone and Joint Staphylococcus aureus Infection. Microbiol Spectr 2023; 11:e0454022. [PMID: 37010409 PMCID: PMC10269916 DOI: 10.1128/spectrum.04540-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/04/2023] [Indexed: 04/04/2023] Open
Abstract
Osteomyelitis is an infection of the bone, associated with an inflammatory process. Imaging plays an important role in establishing the diagnosis and the most appropriate patient management. However, data are lacking regarding the use of preclinical molecular imaging techniques to assess osteomyelitis progression in experimental models. This study aimed to compare structural and molecular imaging to assess disease progression in a mouse model of implant-related bone and joint infections caused by Staphylococcus aureus. In SWISS mice, the right femur was implanted with a resorbable filament impregnated with S. aureus (infected group, n = 10) or sterile culture medium (uninfected group, n = 6). Eight animals (5 infected, 3 uninfected) were analyzed with magnetic resonance imaging (MRI) at 1, 2, and 3 weeks postintervention, and 8 mice were analyzed with [18F]fluorodeoxyglucose (FDG)-positron emission tomography (PET)-computed tomography (CT) at 48 h and at 1, 2, and 3 weeks postintervention. In infected animals, CT showed bone lesion progression, mainly in the distal epiphysis, although some uninfected animals presented evident bone sequestra at 3 weeks. MRI showed a lesion in the articular area that persisted for 3 weeks in infected animals. This lesion was smaller and less evident in the uninfected group. At 48 h postintervention, FDG-PET showed higher joint uptake in the infected group than in the uninfected group (P = 0.025). Over time, the difference between groups increased. These results indicate that FDG-PET imaging was much more sensitive than MRI and CT for differentiating between infection and inflammation at early stages. FDG-PET clearly distinguished between infection and postsurgical bone healing (in uninfected animals) from 48 h to 3 weeks after implantation. IMPORTANCE Our results encourage future investigations on the utility of the model for testing different therapeutic procedures for osteomyelitis.
Collapse
Affiliation(s)
- J. J. Aguilera-Correa
- Departamento de Química en Ciencias Farmacéuticas. Universidad Complutense de Madrid, Madrid, Spain
- CIBERINFEC-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - B. Salinas
- Unidad de Medicina y Cirugía Experimenta, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Salud Mental, Instituto de Salud Carlos III. Madrid, Spain
| | - M. González-Arjona
- Unidad de Medicina y Cirugía Experimenta, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - D. de Pablo
- Servicio de Anatomía Patológica Hospital Clínico San Carlos, Fundación para la Investigación Biomédica HCSC, Madrid, Spain
| | - P. Muñoz
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
- CIBER Enfermedades Respiratorias, CIBERES, Madrid, Spain
| | - E. Bouza
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
- CIBER Enfermedades Respiratorias, CIBERES, Madrid, Spain
| | - M. J. Fernández Aceñero
- Servicio de Anatomía Patológica Hospital Clínico San Carlos, Fundación para la Investigación Biomédica HCSC, Madrid, Spain
| | - J. Esteban
- CIBERINFEC-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Microbiology Department, IIS-Fundacion Jimenez Diaz, UAM, Madrid, Spain
| | - M. Desco
- Unidad de Medicina y Cirugía Experimenta, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Salud Mental, Instituto de Salud Carlos III. Madrid, Spain
| | - L. Cussó
- Unidad de Medicina y Cirugía Experimenta, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Salud Mental, Instituto de Salud Carlos III. Madrid, Spain
| |
Collapse
|
14
|
Mandal S, Tannert A, Ebert C, Guliev RR, Ozegowski Y, Carvalho L, Wildemann B, Eiserloh S, Coldewey SM, Löffler B, Bastião Silva L, Hoerr V, Tuchscherr L, Neugebauer U. Insights into S. aureus-Induced Bone Deformation in a Mouse Model of Chronic Osteomyelitis Using Fluorescence and Raman Imaging. Int J Mol Sci 2023; 24:9762. [PMID: 37298718 PMCID: PMC10254067 DOI: 10.3390/ijms24119762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Osteomyelitis is an infection of the bone that is often difficult to treat and causes a significant healthcare burden. Staphylococcus aureus is the most common pathogen causing osteomyelitis. Osteomyelitis mouse models have been established to gain further insights into the pathogenesis and host response. Here, we use an established S. aureus hematogenous osteomyelitis mouse model to investigate morphological tissue changes and bacterial localization in chronic osteomyelitis with a focus on the pelvis. X-ray imaging was performed to follow the disease progression. Six weeks post infection, when osteomyelitis had manifested itself with a macroscopically visible bone deformation in the pelvis, we used two orthogonal methods, namely fluorescence imaging and label-free Raman spectroscopy, to characterise tissue changes on a microscopic scale and to localise bacteria in different tissue regions. Hematoxylin and eosin as well as Gram staining were performed as a reference method. We could detect all signs of a chronically florid tissue infection with osseous and soft tissue changes as well as with different inflammatory infiltrate patterns. Large lesions dominated in the investigated tissue samples. Bacteria were found to form abscesses and were distributed in high numbers in the lesion, where they could occasionally also be detected intracellularly. In addition, bacteria were found in lower numbers in surrounding muscle tissue and even in lower numbers in trabecular bone tissue. The Raman spectroscopic imaging revealed a metabolic state of the bacteria with reduced activity in agreement with small cell variants found in other studies. In conclusion, we present novel optical methods to characterise bone infections, including inflammatory host tissue reactions and bacterial adaptation.
Collapse
Affiliation(s)
- Shibarjun Mandal
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745 Jena, Germany
| | - Astrid Tannert
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745 Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany (L.T.)
| | - Christina Ebert
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745 Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany (L.T.)
| | - Rustam R. Guliev
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745 Jena, Germany
| | - Yvonne Ozegowski
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany (L.T.)
- Institute for Medical Microbiology, Jena University Hospital, 07747 Jena, Germany
| | - Lina Carvalho
- Institute of Anatomical and Molecular Pathology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Britt Wildemann
- Experimental Trauma Surgery, Jena University Hospital, 07747 Jena, Germany
| | - Simone Eiserloh
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745 Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany (L.T.)
| | - Sina M. Coldewey
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany (L.T.)
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany
| | - Bettina Löffler
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany (L.T.)
- Institute for Medical Microbiology, Jena University Hospital, 07747 Jena, Germany
| | | | - Verena Hoerr
- Institute for Medical Microbiology, Jena University Hospital, 07747 Jena, Germany
- Heart Center Bonn, Department of Internal Medicine II, University Hospital Bonn, 53127 Bonn, Germany
| | - Lorena Tuchscherr
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany (L.T.)
- Institute for Medical Microbiology, Jena University Hospital, 07747 Jena, Germany
| | - Ute Neugebauer
- Leibniz Institute of Photonic Technology (Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research, LPI), 07745 Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany (L.T.)
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
15
|
Zhao Z, Zhang Y, Cheng Y, Li J, Wang W, Huang S, Ma X, Zhang X. Thermosensitive Nanotherapeutics for Localized Photothermal Ablation of MRSA-Infected Osteomyelitis Combined with Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12842-12854. [PMID: 36862542 DOI: 10.1021/acsami.2c23312] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Chronic osteomyelitis is an inflammatory skeletal disease caused by a bacterial infection that affects the periosteum, bone, and bone marrow. Methicillin-resistant Staphylococcus aureus (MRSA) is the most common causative agent. The bacterial biofilm formed on the necrotic bone is a considerable challenge to treating MRSA-infected osteomyelitis. Here, we developed an all-in-one cationic thermosensitive nanotherapeutic (TLCA) for treating MRSA-infected osteomyelitis. The prepared TLCA particles were positively charged and <230 nm in size, which allowed them to diffuse effectively into the biofilm. The positive charges of the nanotherapeutic accurately targeted the biofilm, and it subsequently regulated the drug release under near-infrared (NIR) light irradiation, thereby efficiently exerting the synergistic effect of NIR light-driven photothermal sterilization and chemotherapy. More than 80% of the antibiotics were abruptly released at 50 °C, which dispersed the biofilm by up to 90%. When applied to MRSA-infected osteomyelitis, with a localized temperature of 50 °C induced by 808 nm laser irradiation, it not only eliminated the bacteria and controlled infection but also inhibited the bone tissue inflammatory response, significantly reducing TNF-α, IL-1β, and IL-6 levels. In conclusion, we constructed an all-in-one antimicrobial treatment modality that provides a new and effective strategy for the topical treatment of chronic osteomyelitis.
Collapse
Affiliation(s)
- Zhe Zhao
- Department of Orthopaedics, Tianjin Hospital, No. 406 Jiefangnan Road, Hexi District, Tianjin 300211, China
| | - Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yijie Cheng
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jie Li
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Wenbo Wang
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Siyuan Huang
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xinlong Ma
- Department of Orthopaedics, Tianjin Hospital, No. 406 Jiefangnan Road, Hexi District, Tianjin 300211, China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
16
|
Jia K, Zhang J, Li T, Zhang Y, An J. Comparison of the histopathological characteristics of diffuse sclerosing osteomyelitis of the mandible, chronic suppurative osteomyelitis, and craniofacial fibrous dysplasia. J Oral Pathol Med 2023; 52:91-98. [PMID: 36370060 DOI: 10.1111/jop.13384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 09/27/2022] [Accepted: 11/01/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND There are relatively few reports on the histopathological characteristics of diffuse sclerosing osteomyelitis of the mandible (DSOM), which is difficult to distinguish from chronic suppurative osteomyelitis (CSO) and craniofacial fibrous dysplasia (CFD). This study aimed to summarize and compare the histopathological characteristics of DSOM, CFD, and CSO. MATERIALS AND METHODS In this study, hematoxylin and eosin-stained sections of patients with DSOM, CSO, and CFD at the Peking University Hospital of Stomatology from 2015 to 2020 were retrieved. The histopathological characteristics were summarized, including new bone formation, inflammatory cell infiltration, bone trabecular morphology, osteoclasts, sequestrum, bacterial mass, and calcified spherules, similar to cementicles. The histopathological characteristics of DSOM, CSO, and CFD were compared, and the results were statistically analyzed. RESULTS In total, 50, 13, and 10 patients with DSOM, CSO, and CFD were included in this study, respectively. In terms of new bone formation, both DSOM and CSO showed reactive bone formation (p = 1), whereas CFD mainly showed fiber osteogenesis (p < 0.001). The inflammatory cells of DSOM were mainly lymphocytes and plasma cells, whereas those of CSO were mainly lymphocytes and neutrophils (p < 0.001), and there was usually no inflammatory cell infiltration in the CFD specimens (p < 0.001). DSOM, CSO, and CFD showed irregular bone trabeculae (p = 0.045, p = 0.703) and active osteoclasts (p1 = 0.189, p2 = 0.256). DSOM showed a small amount of bacterial mass but no sequestrum; neither of which was found in CFD (p = 1, p = 1), but it was common in CSO (p = 0.011 and p = 0.025). DSOM and CSO showed smooth and regular basophilic lines (p = 0.308), whereas CFD showed a rough and irregular basophilic line (p < 0.001). CONCLUSIONS The histopathological characteristics of the three diseases were partly similar, but there were evident differences. The main differences are the type of new bone formation, types and distribution of inflammatory cells, and presence of sequestrum and bacterial masses. These differences will help clinicians diagnose DSOM.
Collapse
Affiliation(s)
- Kuankuan Jia
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Jianyun Zhang
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,Department of Oral Pathology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Tiejun Li
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,Department of Oral Pathology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), Beijing, People's Republic of China
| | - Yi Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Jingang An
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| |
Collapse
|
17
|
Staphylococcus aureus Infection Initiates Hypoxia-Mediated Transforming Growth Factor-β1 Upregulation to Trigger Osteomyelitis. mSystems 2022; 7:e0038022. [PMID: 35852344 PMCID: PMC9426532 DOI: 10.1128/msystems.00380-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Little is unknown about the regulatory mechanisms underlying the pathogenesis of osteomyelitis induced by Staphylococcus aureus. Hypoxia-inducible factor-1α (HIF-1α) and transforming growth factor β1 (TGF-β1) were both upregulated in S. aureus-infected MC3T3-E1 cells and osteomyelitis patients. HIF-1α directly targets the hypoxia-responsive elements (HREs) of TGF-β1 mRNA to induce its expression. Silencing HIF-1α and TGF-β1, as well as treatment of hypoxia inhibitor IDF-11774, consistently elevated OPN and RUNX2 expression and alizarin Red S (ARS) and alkaline phosphatase (ALP) staining levels in MC3T3-E1 cells with S. aureus infection. S. aureus infection increased HIF-1α expression and serum TGF-β1 concentration in a mouse model of osteomyelitis. Hypoxia inhibitor IDF-11774 treatment reduced serum levels of interleukin (IL)-6, IL-1β, and C-reactive protein. Upon S. aureus infection, hypoxia was activated to trigger TGF-β1 upregulation through direct targeting of HRE on TGF-β1 mRNA by HIF-1α, eventually leading to osteomyelitis symptoms in terms of osteogenesis and mineralization deficiencies as well as elevated inflammation. This study hereby suggests a novel signaling cascade involving hypoxia/HIF-1α/TGF-β1 in osteomyelitis pathogenesis, which could potentially serve as a target for therapeutic measures. IMPORTANCE The pathogenesis of osteomyelitis induced by Staphylococcus aureus remains unclear. To develop therapeutic approaches for osteomyelitis, it is important to understand the molecular mechanisms of its pathogenesis. Our results suggests that hypoxia/HIF-1α/TGF-β1 signaling is involved in osteomyelitis pathogenesis. Thus, these findings highlight the potential of this signaling components as therapeutic targets for the treatment of osteomyelitis.
Collapse
|
18
|
Radzieta M, Malone M, Ahmad M, Dickson HG, Schwarzer S, Jensen SO, Lavery LA. Metatranscriptome sequencing identifies Escherichia are major contributors to pathogenic functions and biofilm formation in diabetes related foot osteomyelitis. Front Microbiol 2022; 13:956332. [PMID: 35979499 PMCID: PMC9376677 DOI: 10.3389/fmicb.2022.956332] [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: 05/30/2022] [Accepted: 07/11/2022] [Indexed: 11/26/2022] Open
Abstract
Osteomyelitis in the feet of persons with diabetes is clinically challenging and is associated with high rates of amputation. In this study RNA-sequencing was employed to explore microbial metatranscriptomes with a view to understand the relative activity and functions of the pathogen/s responsible for diabetes foot osteomyelitis (DFO). We obtained 25 intraoperative bone specimens from persons with confirmed DFO, observing that Escherichia spp. (7%), Streptomyces spp. (7%), Staphylococcus spp. (6%), Klebsiella spp. (5%) and Proteus spp. (5%) are the most active taxa on average. Data was then subset to examine functions associated with pathogenesis (virulence and toxins), biofilm formation and antimicrobial/multi-drug resistance. Analysis revealed Escherichia spp. are the most active taxa relative to pathogenic functions with K06218 (mRNA interferase relE), K03699 (membrane damaging toxin tlyC) and K03980 (putative peptidoglycan lipid II flippase murJ), K01114 (membrane damaging toxin plc) and K19168 (toxin cptA) being the most prevalent pathogenic associated transcripts. The most abundant transcripts associated with biofilm pathways included components of the biofilm EPS matrix including glycogen synthesis, cellulose synthesis, colonic acid synthesis and flagella synthesis. We further observed enrichment of a key enzyme involved in the biosynthesis of L-rhamnose (K01710 -dTDP-glucose 4,6-dehydratase rfbB, rmlB, rffG) which was present in all but four patients with DFO.
Collapse
Affiliation(s)
- Michael Radzieta
- South West Sydney Limb Preservation and Wound Research, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
- Infectious Diseases and Microbiology, School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Matthew Malone
- South West Sydney Limb Preservation and Wound Research, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
- Infectious Diseases and Microbiology, School of Medicine, Western Sydney University, Sydney, NSW, Australia
- *Correspondence: Matthew Malone
| | - Mehtab Ahmad
- Department of Vascular Surgery, Liverpool Hospital, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
| | - Hugh G. Dickson
- South West Sydney Limb Preservation and Wound Research, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
- South Western Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Saskia Schwarzer
- South West Sydney Limb Preservation and Wound Research, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
- Infectious Diseases and Microbiology, School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Slade O. Jensen
- South West Sydney Limb Preservation and Wound Research, South Western Sydney Local Health District (LHD), Sydney, NSW, Australia
- South Western Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Lawrence A. Lavery
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
19
|
Miek L, Jordan PM, Günther K, Pace S, Beyer T, Kowalak D, Hoerr V, Löffler B, Tuchscherr L, Serhan CN, Gerstmeier J, Werz O. Staphylococcus aureus controls eicosanoid and specialized pro-resolving mediator production via lipoteichoic acid. Immunology 2022; 166:47-67. [PMID: 35143048 PMCID: PMC9426618 DOI: 10.1111/imm.13449] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus causes severe infections associated with inflammation, such as sepsis or osteomyelitis. Inflammatory processes are regulated by distinct lipid mediators (LMs) but how their biosynthetic pathways are orchestrated in S. aureus infections is elusive. We show that S. aureus strikingly not only modulates pro-inflammatory, but also inflammation-resolving LM pathways in murine osteomyelitis and osteoclasts as well as in human monocyte-derived macrophages (MDMs) with different phenotype. Targeted LM metabololipidomics using ultra-performance liquid chromatography-tandem mass spectrometry revealed massive generation of LM with distinct LM signature profiles in acute and chronic phases of S. aureus-induced murine osteomyelitis in vivo. In human MDM, S. aureus elevated cyclooxygenase-2 (COX-2) and microsomal prostaglandin E2 synthase-1 (mPGES-1), but impaired the levels of 15-lipoxygenase-1 (15-LOX-1), with respective changes in LM signature profiles initiated by these enzymes, that is, elevated PGE2 and impaired specialized pro-resolving mediators, along with reduced M2-like phenotypic macrophage markers. The cell wall component, lipoteichoic acid (LTA), mimicked the impact of S. aureus elevating COX-2/mPGES-1 expression via NF-κB and p38 MAPK signalling in MDM, while the impairment of 15-LOX-1 correlates with reduced expression of Lamtor1. In conclusion, S. aureus dictates LM pathways via LTA resulting in a shift from anti-inflammatory M2-like towards pro-inflammatory M1-like LM signature profiles.
Collapse
Affiliation(s)
- Laura Miek
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich‐Schiller‐University JenaJenaGermany
| | - Paul M. Jordan
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich‐Schiller‐University JenaJenaGermany
| | - Kerstin Günther
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich‐Schiller‐University JenaJenaGermany
| | - Simona Pace
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich‐Schiller‐University JenaJenaGermany
| | - Timo Beyer
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich‐Schiller‐University JenaJenaGermany
| | - David Kowalak
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich‐Schiller‐University JenaJenaGermany
| | - Verena Hoerr
- Institute of Medical MicrobiologyJena University HospitalJenaGermany
| | - Bettina Löffler
- Institute of Medical MicrobiologyJena University HospitalJenaGermany
| | - Lorena Tuchscherr
- Institute of Medical MicrobiologyJena University HospitalJenaGermany
| | - Charles N. Serhan
- Department of Anesthesiology, Perioperative and Pain MedicineHarvard Medical SchoolCenter for Experimental Therapeutics and Reperfusion InjuryBrigham and Women’s HospitalBostonMassachusettsUSA
| | - Jana Gerstmeier
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich‐Schiller‐University JenaJenaGermany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal ChemistryInstitute of PharmacyFriedrich‐Schiller‐University JenaJenaGermany
| |
Collapse
|
20
|
Qiu X, Li S, Li X, Xiao Y, Li S, Fen Q, Kang X, Zhen P. Experimental study of β-TCP scaffold loaded with VAN/PLGA microspheres in the treatment of infectious bone defects. Colloids Surf B Biointerfaces 2022; 213:112424. [PMID: 35227993 DOI: 10.1016/j.colsurfb.2022.112424] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 10/19/2022]
Abstract
Antibiotic bone cement filling technology has been widely used in the treatment of infectious bone defects for decades. However, the current treatment requires multiple complicated procedures, which would lead to pain and financial burden for patients. Repairing bone defects and control infection at the same time is the pain spot of orthopaedic area. In this study, we develop a composite scaffold that aiming at effectively repair infectious bone defects simultaneously. Vancomycin hydrochloride(Van) /Poly(lactic-co-glycolic) acid(PLGA) microspheres prepared by double emulsion method were successfully loaded into β-tricalcium phosphate scaffold through electrostatic and physical crosslinking. Full characterization, including mechanical properties, biocompatibility, in vitro release profile and antibacterial properties of the composite scaffolds(CPSFs) were performed. The rabbit osteomyelitis model based on big hole and small hole methods was established. Pharmacodynamics study, including the local bacteriostatic and osteogenic ability were evaluated by X-ray, Micro-CT and histopathology at 4 months after surgery. These findings indicate that a reliable rabbit model of local bone defect infection successfully established by big hole approach. The CPSFs with significant histocompatibility and biocompatibility could sustained release vancomycin for extended duration. It exhibited great application potential in clinical aim at the indication of local infectious bone defects.
Collapse
Affiliation(s)
- Xiaoming Qiu
- Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Songkai Li
- Department of the orthopaedic centre, The 940th Hospital of Logistics Support Force of PLA, Lanzhou 730000, China
| | - Xun Li
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yating Xiao
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Shengtang Li
- Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Qiangsheng Fen
- Department of the orthopaedic centre, The 940th Hospital of Logistics Support Force of PLA, Lanzhou 730000, China
| | - Xuewen Kang
- Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Ping Zhen
- Lanzhou University Second Hospital, Lanzhou 730000, China; Department of the orthopaedic centre, The 940th Hospital of Logistics Support Force of PLA, Lanzhou 730000, China
| |
Collapse
|
21
|
Billings C, Anderson DE. Role of Animal Models to Advance Research of Bacterial Osteomyelitis. Front Vet Sci 2022; 9:879630. [PMID: 35558882 PMCID: PMC9087578 DOI: 10.3389/fvets.2022.879630] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Osteomyelitis is an inflammatory bone disease typically caused by infectious microorganisms, often bacteria, which causes progressive bone destruction and loss. The most common bacteria associated with chronic osteomyelitis is Staphylococcus aureus. The incidence of osteomyelitis in the United States is estimated to be upwards of 50,000 cases annually and places a significant burden upon the healthcare system. There are three general categories of osteomyelitis: hematogenous; secondary to spread from a contiguous focus of infection, often from trauma or implanted medical devices and materials; and secondary to vascular disease, often a result of diabetic foot ulcers. Independent of the route of infection, osteomyelitis is often challenging to diagnose and treat, and the effect on the patient's quality of life is significant. Therapy for osteomyelitis varies based on category and clinical variables in each case. Therapeutic strategies are typically reliant upon protracted antimicrobial therapy and surgical interventions. Therapy is most successful when intensive and initiated early, although infection may recur months to years later. Also, treatment is accompanied by risks such as systemic toxicity, selection for antimicrobial drug resistance from prolonged antimicrobial use, and loss of form or function of the affected area due to radical surgical debridement or implant removal. The challenges of diagnosis and successful treatment, as well as the negative impacts on patient's quality of life, exemplify the need for improved strategies to combat bacterial osteomyelitis. There are many in vitro and in vivo investigations aimed toward better understanding of the pathophysiology of bacterial osteomyelitis, as well as improved diagnostic and therapeutic strategies. Here, we review the role of animal models utilized for the study of bacterial osteomyelitis and their critically important role in understanding and improving the management of bacterial osteomyelitis.
Collapse
|
22
|
Dietrich O, Heinz A, Goldmann O, Geffers R, Beineke A, Hiller K, Saliba AE, Medina E. Dysregulated Immunometabolism Is Associated with the Generation of Myeloid-Derived Suppressor Cells in Staphylococcus aureus Chronic Infection. J Innate Immun 2021; 14:257-274. [PMID: 34763332 DOI: 10.1159/000519306] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/23/2021] [Indexed: 11/19/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a compendium of immature myeloid cells that exhibit potent T-cell suppressive capacity and expand during pathological conditions such as cancer and chronic infections. Although well-characterized in cancer, the physiology of MDSCs in the infection setting remains enigmatic. Here, we integrated single-cell RNA sequencing (scRNA-seq) and functional metabolic profiling to gain deeper insights into the factors governing the generation and maintenance of MDSCs in chronic Staphylococcus aureus infection. We found that MDSCs originate not only in the bone marrow but also at extramedullary sites in S. aureus-infected mice. scRNA-seq showed that infection-driven MDSCs encompass a spectrum of myeloid precursors in different stages of differentiation, ranging from promyelocytes to mature neutrophils. Furthermore, the scRNA-seq analysis has also uncovered valuable phenotypic markers to distinguish mature myeloid cells from immature MDSCs. Metabolic profiling indicates that MDSCs exhibit high glycolytic activity and high glucose consumption rates, which are required for undergoing terminal maturation. However, rapid glucose consumption by MDSCs added to infection-induced perturbations in the glucose supplies in infected mice hinders the terminal maturation of MDSCs and promotes their accumulation in an immature stage. In a proof-of-concept in vivo experiment, we demonstrate the beneficial effect of increasing glucose availability in promoting MDSC terminal differentiation in infected mice. Our results provide valuable information of how metabolic alterations induced by infection influence reprogramming and differentiation of MDSCs.
Collapse
Affiliation(s)
- Oliver Dietrich
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Wuerzburg, Germany
| | - Alexander Heinz
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany.,Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Wuerzburg, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
23
|
Roper PM, Eichelberger KR, Cox L, O’Connor L, Shao C, Ford CA, Fritz SA, Cassat JE, Veis DJ. Contemporary Clinical Isolates of Staphylococcus aureus from Pediatric Osteomyelitis Patients Display Unique Characteristics in a Mouse Model of Hematogenous Osteomyelitis. Infect Immun 2021; 89:e0018021. [PMID: 34097469 PMCID: PMC8445171 DOI: 10.1128/iai.00180-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/25/2021] [Indexed: 11/20/2022] Open
Abstract
Osteomyelitis can result from the direct inoculation of pathogens into bone during injury or surgery or from spread via the bloodstream, a condition called hematogenous osteomyelitis (HOM). HOM disproportionally affects children, and more than half of cases are caused by Staphylococcus aureus. Laboratory models of osteomyelitis mostly utilize direct injection of bacteria into the bone or implantation of foreign material and therefore do not directly interrogate the pathogenesis of pediatric hematogenous osteomyelitis. In this study, we inoculated mice intravenously and characterized the resultant musculoskeletal infections using two strains isolated from adults (USA300-LAC and NRS384) and five new methicillin-resistant S. aureus isolates from pediatric osteomyelitis patients. All strains were capable of creating stable infections over 5 weeks, although the incidence varied. Micro-computed tomography (microCT) analysis demonstrated decreases in the trabecular bone volume fraction but little effect on bone cortices. Histological assessment revealed differences in the precise focus of musculoskeletal infection, with various mixtures of bone-centered osteomyelitis and joint-centered septic arthritis. Whole-genome sequencing of three new isolates demonstrated distinct strains, two within the USA300 lineage and one USA100 isolate. Interestingly, this USA100 isolate showed a distinct predilection for septic arthritis compared to the other isolates tested, including NRS384 and LAC, which more frequently led to osteomyelitis or mixed bone and joint infections. Collectively, these data outline the feasibility of using pediatric osteomyelitis clinical isolates to study the pathogenesis of HOM in murine models and lay the groundwork for future studies investigating strain-dependent differences in musculoskeletal infection.
Collapse
Affiliation(s)
- Philip M. Roper
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Kara R. Eichelberger
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Linda Cox
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Luke O’Connor
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Christine Shao
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Caleb A. Ford
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Stephanie A. Fritz
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - James E. Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Deborah J. Veis
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Shriners Hospitals for Children, Saint Louis, Missouri, USA
| |
Collapse
|
24
|
Alstrup AKO, Jensen SB, Nielsen OL, Jødal L, Afzelius P. Preclinical Testing of Radiopharmaceuticals for the Detection and Characterization of Osteomyelitis: Experiences from a Porcine Model. Molecules 2021; 26:molecules26144221. [PMID: 34299496 PMCID: PMC8305428 DOI: 10.3390/molecules26144221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/30/2021] [Accepted: 07/10/2021] [Indexed: 12/01/2022] Open
Abstract
The development of new and better radioactive tracers capable of detecting and characterizing osteomyelitis is an ongoing process, mainly because available tracers lack selectivity towards osteomyelitis. An integrated part of developing new tracers is the performance of in vivo tests using appropriate animal models. The available animal models for osteomyelitis are also far from ideal. Therefore, developing improved animal osteomyelitis models is as important as developing new radioactive tracers. We recently published a review on radioactive tracers. In this review, we only present and discuss osteomyelitis models. Three ethical aspects (3R) are essential when exposing experimental animals to infections. Thus, we should perform experiments in vitro rather than in vivo (Replacement), use as few animals as possible (Reduction), and impose as little pain on the animal as possible (Refinement). The gain for humans should by far exceed the disadvantages for the individual experimental animal. To this end, the translational value of animal experiments is crucial. We therefore need a robust and well-characterized animal model to evaluate new osteomyelitis tracers to be sure that unpredicted variation in the animal model does not lead to a misinterpretation of the tracer behavior. In this review, we focus on how the development of radioactive tracers relies heavily on the selection of a reliable animal model, and we base the discussions on our own experience with a porcine model.
Collapse
Affiliation(s)
- Aage Kristian Olsen Alstrup
- Department of Nuclear Medicine & PET, Aarhus University Hospital, DK-8200 Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, DK-8200 Aarhus, Denmark
- Correspondence: ; Tel.: +45-22899285
| | - Svend Borup Jensen
- Department of Nuclear Medicine, Aalborg University Hospital, DK-9000 Aalborg, Denmark; (S.B.J.); (L.J.)
- Department of Chemistry and Biosciences, Aalborg University, DK-9220 Aalborg, Denmark
| | - Ole Lerberg Nielsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, DK-1165 Copenhagen, Denmark;
| | - Lars Jødal
- Department of Nuclear Medicine, Aalborg University Hospital, DK-9000 Aalborg, Denmark; (S.B.J.); (L.J.)
| | - Pia Afzelius
- Zealand Hospital, Køge, Copenhagen University Hospital, DK-4600 Køge, Denmark;
| |
Collapse
|
25
|
Staphylococcus epidermidis small colony variants, clinically significant quiescent threats for patients with prosthetic joint infection. Microbes Infect 2021; 23:104854. [PMID: 34214690 DOI: 10.1016/j.micinf.2021.104854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 11/23/2022]
|
26
|
Choi YS, Ham DS, Lim JY, Lee YK. Validation of the Osteomyelitis Induced by Methicillin-Resistant Staphylococcus aureus (MRSA) on Rat Model with Calvaria Defect. Tissue Eng Regen Med 2021; 18:671-683. [PMID: 34165776 DOI: 10.1007/s13770-021-00340-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Osteomyelitis resulting from bacterial strains, such as methicillin-resistant Staphylococcus aureus (MRSA) that are resistant to multiple drugs, brings further clinical challenges. There is currently no model of osteomyelitis induced by MRSA using rats with calvaria defects. So, We induced osteomyelitis in rat models with the calvaria bone defect. METHODS The rats were randomly divided into six groups according to inoculation dose levels, which ranged from 6 × 100 to 6 × 105 CFU/5 µl. Bone tissues were retrieved from all rats used in the study and assessed using histology, microbiology, and radiobiology 4 weeks after surgery to evaluate the relationship between inoculation dose and infectivity. RESULTS In Histological results, high levels of inflammatory responses, bone necrosis, and bacteria were observed in treatment groups G3 to G5. In IHC staining, high levels of cox-2 expression were observed in treatment groups G3. Microbiological observations also indicated that significantly higher numbers of CFUs were found in G3 to G5. In radiography results, the bone mineral density in G3 to G5 was significantly higher than in the control group, G1, and G2. Our results indicate that an inoculating dose of 6 × 103 CFU/5 μl is sufficient to induce the development of osteomyelitis in rat models. CONCLUSION This study suggests that the minimum dose (6 × 103 CFU/5 µl) can induce osteomyelitis in calvaria rat model. This can offer information and ability of more accurately modeling osteomyelitis and simulating the challenge of osteomyelitis treat.
Collapse
Affiliation(s)
- Young Suk Choi
- Department of Biology, Soonchunhyang University, Soonchunhyang-ro, Sinchang-myeon, Asan-si, Chungcheongnam-do, 31538, Republic of Korea.,Department of Orthopedic Surgery, Soonchunhyang University Bucheon Hospital, 4 Jung-Dong, Wonmi-Gu, Bucheon-Si, Gyeonggi-Do, 14584, Republic of Korea
| | - Dae Sung Ham
- Department of Orthopedic Surgery, Soonchunhyang University Bucheon Hospital, 4 Jung-Dong, Wonmi-Gu, Bucheon-Si, Gyeonggi-Do, 14584, Republic of Korea.,Department of Medical Sciences, Soonchunhyang University, Soonchunhyang-ro, Sinchang-myeon, Asan-si, Chungcheongnam-do, 31538, Republic of Korea
| | - Ji Yun Lim
- Department of Orthopedic Surgery, Soonchunhyang University Bucheon Hospital, 4 Jung-Dong, Wonmi-Gu, Bucheon-Si, Gyeonggi-Do, 14584, Republic of Korea.,Department of Medical Sciences, Soonchunhyang University, Soonchunhyang-ro, Sinchang-myeon, Asan-si, Chungcheongnam-do, 31538, Republic of Korea
| | - Young Koo Lee
- Department of Orthopedic Surgery, Soonchunhyang University Bucheon Hospital, 4 Jung-Dong, Wonmi-Gu, Bucheon-Si, Gyeonggi-Do, 14584, Republic of Korea.
| |
Collapse
|
27
|
Wang J, Wang L. Novel therapeutic interventions towards improved management of septic arthritis. BMC Musculoskelet Disord 2021; 22:530. [PMID: 34107951 PMCID: PMC8191206 DOI: 10.1186/s12891-021-04383-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/18/2021] [Indexed: 01/19/2023] Open
Abstract
Septic arthritis (SA) represents a medical emergency that needs immediate diagnosis and urgent treatment. Despite aggressive treatment and rapid diagnosis of the causative agent, the mortality and lifelong disability, associated with septic arthritis remain high as close to 11%. Moreover, with the rise in drug resistance, the rates of failure of conventional antibiotic therapy have also increased. Among the etiological agents frequently isolated from cases of septic arthritis, Staphylococcus aureus emerges as a dominating pathogen, and to worsen, the rise in methicillin-resistant S. aureus (MRSA) isolates in bone and joint infections is worrisome. MRSA associated cases of septic arthritis exhibit higher mortality, longer hospital stay, and higher treatment failure with poorer clinical outcomes as compared to cases caused by the sensitive strain i.e methicillin-sensitive S. aureus (MSSA). In addition to this, equal or even greater damage is imposed by the exacerbated immune response mounted by the patient’s body in a futile attempt to eradicate the bacteria. The antibiotic therapy may not be sufficient enough to control the progression of damage to the joint involved thus, adding to higher mortality and disability rates despite the prompt and timely start of treatment. This situation implies that efforts and focus towards studying/understanding new strategies for improved management of sepsis arthritis is prudent and worth exploring. The review article aims to give a complete insight into the new therapeutic approaches studied by workers lately in this field. To the best of our knowledge studies highlighting the novel therapeutic strategies against septic arthritis are limited in the literature, although articles on pathogenic mechanism and choice of antibiotics for therapy, current treatment algorithms followed have been discussed by workers in the past. The present study presents and discusses the new alternative approaches, their mechanism of action, proof of concept, and work done so far towards their clinical success. This will surely help to enlighten the researchers with comprehensive knowledge of the new interventions that can be used as an adjunct therapy along with conventional treatment protocol for improved success rates.
Collapse
Affiliation(s)
- Jian Wang
- Department of Nursing, The Third Hospital of Jinan, Shandong Province, Jinan, 250132, China.
| | - Liucai Wang
- Hand and Foot Surgery, Shandong Provincial Hospital, Jinan, 250000, China
| |
Collapse
|
28
|
Avican K, Aldahdooh J, Togninalli M, Mahmud AKMF, Tang J, Borgwardt KM, Rhen M, Fällman M. RNA atlas of human bacterial pathogens uncovers stress dynamics linked to infection. Nat Commun 2021; 12:3282. [PMID: 34078900 PMCID: PMC8172932 DOI: 10.1038/s41467-021-23588-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 05/05/2021] [Indexed: 11/25/2022] Open
Abstract
Bacterial processes necessary for adaption to stressful host environments are potential targets for new antimicrobials. Here, we report large-scale transcriptomic analyses of 32 human bacterial pathogens grown under 11 stress conditions mimicking human host environments. The potential relevance of the in vitro stress conditions and responses is supported by comparisons with available in vivo transcriptomes of clinically important pathogens. Calculation of a probability score enables comparative cross-microbial analyses of the stress responses, revealing common and unique regulatory responses to different stresses, as well as overlapping processes participating in different stress responses. We identify conserved and species-specific 'universal stress responders', that is, genes showing altered expression in multiple stress conditions. Non-coding RNAs are involved in a substantial proportion of the responses. The data are collected in a freely available, interactive online resource (PATHOgenex).
Collapse
Affiliation(s)
- Kemal Avican
- Department of Molecular Biology, Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden.
| | - Jehad Aldahdooh
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Matteo Togninalli
- Department for Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Swiss Institute for Bioinformatics, Lausanne, Switzerland
| | - A K M Firoj Mahmud
- Department of Molecular Biology, Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Jing Tang
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Karsten M Borgwardt
- Department for Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Swiss Institute for Bioinformatics, Lausanne, Switzerland
| | - Mikael Rhen
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Stockholm, Sweden
| | - Maria Fällman
- Department of Molecular Biology, Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden.
| |
Collapse
|
29
|
Arias SL, Brito IL. Biophysical determinants of biofilm formation in the gut. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 18:100275. [PMID: 34504987 PMCID: PMC8423371 DOI: 10.1016/j.cobme.2021.100275] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The gastrointestinal (GI) tract harbors the most complex microbial ecosystem in the human body. The mucosal layer that covers the GI tract serves as a polymer-based defensive barrier that prevents the microbiome from reaching the epithelium and disseminating inside the body. Colonization of the mucus may result in the formation of structured polymicrobial communities or biofilms, a hallmark in pathologies such as colorectal cancer, inflammatory bowel disease, and chronic gut wounds. However, the mechanisms by which multispecies biofilms establish on the gut mucosa is unknown. Whether mucus-associated biofilms exist as part of a healthy mucosal barrier is still debated. Here, we discuss the impact that diet and microbial-derived polymers has on mucus structure and microcolony formation and highlight relevant biophysical forces that further shape nascent biofilms.
Collapse
Affiliation(s)
- Sandra L Arias
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850
| | - Ilana L Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850
| |
Collapse
|
30
|
mRNA Transcriptome Analysis of Bone in a Mouse Model of Implant-Associated Staphylococcus aureus Osteomyelitis. Infect Immun 2021; 89:IAI.00814-20. [PMID: 33619031 DOI: 10.1128/iai.00814-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/02/2021] [Indexed: 02/06/2023] Open
Abstract
To investigate the molecular pathogenesis of bone with osteomyelitis, we developed implant-associated osteomyelitis (IAOM) models in mice. An orthopedic stainless pin was surgically placed in the right femoral midshaft of mice, followed by an inoculation of Staphylococcus aureus into the medullary cavity. Typical characteristics of IAOM, like periosteal reaction and intraosseous abscess, occurred by day 14 postinfection. By day 28 postinfection, necrotic abscess, sequestrum formation, and deformity of the whole femur were observed. Transcriptional analysis identified 101 and 1,702 differentially expressed genes (DEGs) between groups by days 3 and 14 postinfection, respectively. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed the enrichment of pathways in response to the bacterium, receptor-ligand activity, and chemokine signaling by day 3 postinfection. However, by day 14 postinfection, the enrichment switched to angiogenesis, positive regulation of cell motility and migration, skeletal system development, and cytokine-cytokine receptor interaction. Furthermore, protein-protein interaction network analysis identified 4 cytokines (interleukin 6 [IL-6], Cxcl10, gamma interferon [IFN-γ], and Cxcl9) associated with IAOM at an early stage of infection. Overall, as the pathological changes in this mouse model were consistent with those in human IAOM, our model may be used to investigate the mechanism and treatment of IAOM. Furthermore, the data for transcriptome sequencing and bioinformatic analysis will be an important resource for dissecting the molecular pathogenesis of bone with IAOM.
Collapse
|
31
|
Direct interactions between cationic liposomes and bacterial cells ameliorate the systemic treatment of invasive multidrug-resistant Staphylococcus aureus infections. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 34:102382. [PMID: 33771706 DOI: 10.1016/j.nano.2021.102382] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 01/14/2021] [Accepted: 03/03/2021] [Indexed: 12/19/2022]
Abstract
Invasive infections caused by antibiotic-resistant Staphylococcus aureus have posed a great threat to human health. To tackle this problem, a cationic liposomal Curcumin (C-LS/Cur) was developed and its effect against antibiotic-resistant S. aureus was investigated in this study. As expected, C-LS/Cur exhibited greater bactericidal capacity compared with its counterparts, probably because the negatively charged S. aureus favors electrostatic interactions rather than intercalation with cationic liposomal vesicles at the beginning of endocytic process, thereby effectively delivering Cur to its targets. We confirmed this hypothesis by monitoring zeta potential variation, collecting visual evidences through CLSM, FCM and TEM, and determining binding kinetics by BLI. Moreover, an excellent therapeutic efficacy of C-LS/Cur against invasive murine infection was also observed, which was due to the enhanced accumulation and retention in the targets. Therefore, cationic liposomes have great potential for the clinical application in the treatment of invasive antibiotic-resistant S. aureus infections.
Collapse
|
32
|
Nielsen OL, Mellergaard M, Frees D, Larsen MN, Skov S, Olsen LH, Reimann MJ. A porcine model of subcutaneous Staphylococcus aureus infection: a pilot study. APMIS 2021; 130:359-370. [PMID: 33644910 DOI: 10.1111/apm.13101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/18/2020] [Indexed: 11/30/2022]
Abstract
In this descriptive pilot study, we aim to establish a porcine Staphylococcus aureus skin infection model by subcutaneous injection (s.c.) of the porcine S54F9 S. aureus strain in the groin area. Six pigs were used in the study: Five pigs were injected with S. aureus, inocula ranging from 7 × 103 to 5 × 107 colony-forming units per kg bodyweight; one pig was injected with saline exclusively. Lesions were recorded up to 6 days postinoculation using clinical evaluation, ultrasound evaluation, microbiology, flow cytometry, and pathology. Inoculation gave rise to lesions ranging from localized skin infection, that is, minute histological changes, intracellular infection, and macroscopic abscess formation with sequestration of soft tissue, to generalized infection and development of disseminated intravascular coagulation necessitating euthanasia only 10 h after inoculation. Ultrasound assessment of maximum width and characteristics was not able to disclose the progress of the local infection. Flow cytometry and immunohistochemistry revealed the participation of γδT cells in the immune response. In conclusion, we did see a graded inflammatory response associated with the dose of s.c. inoculated bacteria, which may be useful for studying, in particular, the interaction of bacteria and inflammatory mononuclear cell populations. It needs to be investigated if the model is discriminatory and robust.
Collapse
Affiliation(s)
- Ole Lerberg Nielsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Maiken Mellergaard
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Dorte Frees
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Maria Nygaard Larsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Søren Skov
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Lisbeth Høier Olsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Maria Josefine Reimann
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
33
|
Roux KM, Cobb LH, Seitz MA, Priddy LB. Innovations in osteomyelitis research: A review of animal models. Animal Model Exp Med 2021; 4:59-70. [PMID: 33738438 PMCID: PMC7954837 DOI: 10.1002/ame2.12149] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Infection of bone tissue, or osteomyelitis, has become a growing concern in modern healthcare due in no small part to a rise in antibiotic resistance among bacteria, notably Staphylococcus aureus. The current standard of care involves aggressive, prolonged antibiotic therapy combined with surgical debridement of infected tissues. While this treatment may be sufficient for resolving a portion of cases, recurrences of the infection and associated risks including toxicity with long-term antibiotic usage have been reported. Therefore, there exists a need to produce safer, more efficacious options of treatment for osteomyelitis. In order to test treatment regimens, animal models that closely mimic the clinical condition and allow for accurate evaluation of therapeutics are necessary. Establishing a model that replicates features of osteomyelitis in humans continues to be a challenge to scientists, as there are many variables involved, including choosing an appropriate species and method to establish infection. This review addresses the refinement of animal models of osteomyelitis to reflect the clinical disease and test prospective therapeutics. The aim of this review is to explore studies regarding the use of animals for osteomyelitis therapeutics research and encourage further development of such animal models for the translation of results from the animal experiment to human medicine.
Collapse
Affiliation(s)
- Kylie M. Roux
- College of Veterinary MedicineMississippi State UniversityMississippi StateMSUSA
| | - Leah H. Cobb
- Department of Agricultural and Biological EngineeringMississippi State UniversityMississippi StateMSUSA
| | - Marc A. Seitz
- College of Veterinary MedicineMississippi State UniversityMississippi StateMSUSA
| | - Lauren B. Priddy
- Department of Agricultural and Biological EngineeringMississippi State UniversityMississippi StateMSUSA
| |
Collapse
|
34
|
Gimza BD, Cassat JE. Mechanisms of Antibiotic Failure During Staphylococcus aureus Osteomyelitis. Front Immunol 2021; 12:638085. [PMID: 33643322 PMCID: PMC7907425 DOI: 10.3389/fimmu.2021.638085] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus is a highly successful Gram-positive pathogen capable of causing both superficial and invasive, life-threatening diseases. Of the invasive disease manifestations, osteomyelitis or infection of bone, is one of the most prevalent, with S. aureus serving as the most common etiologic agent. Treatment of osteomyelitis is arduous, and is made more difficult by the widespread emergence of antimicrobial resistant strains, the capacity of staphylococci to exhibit tolerance to antibiotics despite originating from a genetically susceptible background, and the significant bone remodeling and destruction that accompanies infection. As a result, there is a need for a better understanding of the factors that lead to antibiotic failure in invasive staphylococcal infections such as osteomyelitis. In this review article, we discuss the different non-resistance mechanisms of antibiotic failure in S. aureus. We focus on how bacterial niche and destructive tissue remodeling impact antibiotic efficacy, the significance of biofilm formation in promoting antibiotic tolerance and persister cell formation, metabolically quiescent small colony variants (SCVs), and potential antibiotic-protected reservoirs within the substructure of bone.
Collapse
Affiliation(s)
- Brittney D Gimza
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James E Cassat
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
35
|
Yamashita J, Sawa N, Sawa Y, Miyazono S. Effect of bisphosphonates on healing of tooth extraction wounds in infectious osteomyelitis of the jaw. Bone 2021; 143:115611. [PMID: 32829042 DOI: 10.1016/j.bone.2020.115611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Antiresorptive agent-related osteonecrosis of the jaw (ARONJ) and infectious osteomyelitis of the jaw (OMJ) in antiresorptive-naïve patients are different disease entities. Although osteoclast inhibition is at the center of the pathogenesis of ARONJ, the role of osteoclast inhibition in infectious OMJ is unknown. The objective of this study was to determine the effect of bisphosphonate osteoclast inhibition in infectious OMJ. METHODS Osteomyelitis was induced in mice by S. aureus inoculation. The establishment of OMJ was verified by the culture of bone marrow samples obtained from the mandible. Infected animals received either zoledronic acid (ZA) or saline starting at week-2. Treated animals along with non-infected animals underwent tooth extractions at week-4 post-infection. Healing was assessed every week using in vivo micro-computed tomography and intraoral photos. Animals were euthanized at week-8 and cervical lymph nodes were assessed for lymphatic and blood vessels. RESULTS Tooth extraction wounds did not heal in animals with OMJ. These wounds were characterized by incomplete soft tissue coverage, sporadic bone fill in the sockets, and inflammatory cell accumulation in the connective tissue at 4 weeks after tooth extractions. Conversely, the majority of tooth extraction wounds in the infected animals treated with ZA had improved healing with better bone fill than even non-infected control animals. Consistently, atrophic lymphatic vessels were noted in the draining lymph nodes in animals with OMJ. However, infected animals treated with ZA had lymphatic vessels that were unaltered and showed a similar appearance to those in non-infected control animals. CONCLUSION ZA treatment promoted wound healing in the jaw with infectious osteomyelitis. Clearly, antiresorptive therapy is contraindicated in patients with ARONJ. However, our finding suggests that osteoclast inhibition is potentially an effectual remedy for infectious OMJ in antiresorptive-naïve patients.
Collapse
Affiliation(s)
- Junro Yamashita
- Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan; Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan.
| | - Naruhiko Sawa
- Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan; Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan
| | - Yoshihiko Sawa
- Department of Oral Function and Anatomy, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shoji Miyazono
- Department of Oral Rehabilitation, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
36
|
Chen H, Yang H, Weir MD, Schneider A, Ren K, Homayounfar N, Oates TW, Zhang K, Liu J, Hu T, Xu HHK. An antibacterial and injectable calcium phosphate scaffold delivering human periodontal ligament stem cells for bone tissue engineering. RSC Adv 2020; 10:40157-40170. [PMID: 35520873 PMCID: PMC9057516 DOI: 10.1039/d0ra06873j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023] Open
Abstract
Osteomyelitis and post-operative infections are major problems in orthopedic, dental and craniofacial surgeries. It is highly desirable for a tissue engineering construct to kill bacteria, while simultaneously delivering stem cells and enhancing cell function and tissue regeneration. The objectives of this study were to: (1) develop a novel injectable calcium phosphate cement (CPC) scaffold containing antibiotic ornidazole (ORZ) while encapsulating human periodontal ligament stem cells (hPDLSCs), and (2) investigate the inhibition efficacy against Staphylococcus aureus (S. aureus) and the promotion of hPDLSC function for osteogenesis for the first time. ORZ was incorporated into a CPC-chitosan scaffold. hPDLSCs were encapsulated in alginate microbeads (denoted hPDLSCbeads). The ORZ-loaded CPCC+hPDLSCbeads scaffold was fully injectable, and had a flexural strength of 3.50 ± 0.92 MPa and an elastic modulus of 1.30 ± 0.45 GPa, matching those of natural cancellous bone. With 6 days of sustained ORZ release, the CPCC+10ORZ (10% ORZ) scaffold had strong antibacterial effects on S. aureus, with an inhibition zone of 12.47 ± 1.01 mm. No colonies were observed in the CPCC+10ORZ group from 3 to 7 days. ORZ-containing scaffolds were biocompatible with hPDLSCs. CPCC+10ORZ+hPDLSCbeads scaffold with osteogenic medium had 2.4-fold increase in alkaline phosphatase (ALP) activity and bone mineral synthesis by hPDLSCs, as compared to the control group (p < 0.05). In conclusion, the novel antibacterial construct with stem cell delivery had injectability, good strength, strong antibacterial effects and biocompatibility, supporting osteogenic differentiation and bone mineral synthesis of hPDLSCs. The injectable and mechanically-strong CPCC+10ORZ+hPDLSCbeads construct has great potential for treating bone infections and promoting bone regeneration.
Collapse
Affiliation(s)
- Hong Chen
- Department of Endodontics, College of Stomatological, Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education Chongqing China
- State Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, National Clinical Research Centre for Oral Diseases, Sichuan University Chengdu China
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
| | - Hui Yang
- State Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, National Clinical Research Centre for Oral Diseases, Sichuan University Chengdu China
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry Baltimore USA
- Member, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine Baltimore MD 21201 USA
| | - Ke Ren
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, University of Maryland Baltimore MD 21201 USA
| | - Negar Homayounfar
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
| | - Thomas W Oates
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
| | - Ke Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University Beijing China
| | - Jin Liu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
- Key Laboratory of Shannxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University Xi'an Shannxi China
| | - Tao Hu
- State Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, National Clinical Research Centre for Oral Diseases, Sichuan University Chengdu China
| | - Hockin H K Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School Baltimore MD 21201 USA
- Member, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine Baltimore MD 21201 USA
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine Baltimore MD 21201 USA
| |
Collapse
|
37
|
Three-Dimensional In Vitro Staphylococcus aureus Abscess Communities Display Antibiotic Tolerance and Protection from Neutrophil Clearance. Infect Immun 2020; 88:IAI.00293-20. [PMID: 32817328 DOI: 10.1128/iai.00293-20] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus is a prominent human pathogen in bone and soft-tissue infections. Pathophysiology involves abscess formation, which consists of central staphylococcal abscess communities (SACs), surrounded by a fibrin pseudocapsule and infiltrating immune cells. Protection against the ingress of immune cells such as neutrophils, or tolerance to antibiotics, remains largely unknown for SACs and is limited by the lack of availability of in vitro models. We describe a three-dimensional in vitro model of SACs grown in a human plasma-supplemented collagen gel. The in vitro SACs reached their maximum size by 24 h and elaborated a fibrin pseudocapsule, as confirmed by electron and immunofluorescence microscopy. The in vitro SACs tolerated 100× the MIC of gentamicin alone and in combination with rifampin, while planktonic controls and mechanically dispersed SACs were efficiently killed. To simulate a host response, SACs were exposed to differentiated PLB-985 neutrophil-like (dPLB) cells and to primary human neutrophils at an early stage of SAC formation or after maturation at 24 h. Both cell types were unable to clear mature in vitro SACs, but dPLB cells prevented SAC growth upon early exposure before pseudocapsule maturation. Neutrophil exposure after plasmin pretreatment of the SACs resulted in a significant decrease in the number of bacteria within the SACs. The in vitro SAC model mimics key in vivo features, offers a new tool to study host-pathogen interactions and drug efficacy assessment, and has revealed the functionality of the S. aureus pseudocapsule in protecting the bacteria from host phagocytic responses and antibiotics.
Collapse
|
38
|
Identification of a Novel LysR-Type Transcriptional Regulator in Staphylococcus aureus That Is Crucial for Secondary Tissue Colonization during Metastatic Bloodstream Infection. mBio 2020; 11:mBio.01646-20. [PMID: 32843554 PMCID: PMC7448277 DOI: 10.1128/mbio.01646-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Staphylococcus aureus is an important pathogen that can disseminate via the bloodstream and establish metastatic infections in distant organs. To achieve a better understanding of the bacterial factors facilitating the development of these metastatic infections, we used in this study a Staphylococcus aureus transposon mutant library in a murine model of intravenous infection, where bacteria first colonize the liver as the primary infection site and subsequently progress to secondary sites such as the kidney and bones. We identified a novel LysR-type transcriptional regulator (LTTR), which was specifically required by S. aureus for efficient colonization of secondary organs. We also determined the transcriptional activation as well as the regulon of LTTR, which suggests that this regulator is involved in the metabolic adaptation of S. aureus to the host microenvironment found in secondary infection sites. Staphylococcus aureus is a common cause of bacteremia that can lead to severe complications once the bacteria exit the bloodstream and establish infection in secondary organs. Despite its clinical relevance, little is known about the bacterial factors facilitating the development of these metastatic infections. Here, we used an S. aureus transposon mutant library coupled to transposon insertion sequencing (Tn-Seq) to identify genes that are critical for efficient bacterial colonization of secondary organs in a murine model of metastatic bloodstream infection. Our transposon screen identified a LysR-type transcriptional regulator (LTTR), which was required for efficient colonization of secondary organs such as the kidneys in infected mice. The critical role of LTTR in secondary organ colonization was confirmed using an isogenic mutant deficient in the expression of LTTR. To identify the set of genes controlled by LTTR, we used an S. aureus strain carrying the LTTR gene in an inducible expression plasmid. Gene expression analysis upon induction of LTTR showed increased transcription of genes involved in branched-chain amino acid biosynthesis, a methionine sulfoxide reductase, and a copper transporter as well as decreased transcription of genes coding for urease and components of pyrimidine nucleotides. Furthermore, we show that transcription of LTTR is repressed by glucose, is induced under microaerobic conditions, and required trace amounts of copper ions. Our data thus pinpoints LTTR as an important element that enables a rapid adaptation of S. aureus to the changing host microenvironment.
Collapse
|
39
|
Bonifacius A, Goldmann O, Floess S, Holtfreter S, Robert PA, Nordengrün M, Kruse F, Lochner M, Falk CS, Schmitz I, Bröker BM, Medina E, Huehn J. Staphylococcus aureus Alpha-Toxin Limits Type 1 While Fostering Type 3 Immune Responses. Front Immunol 2020; 11:1579. [PMID: 32849537 PMCID: PMC7427519 DOI: 10.3389/fimmu.2020.01579] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 06/15/2020] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus can cause life-threatening diseases, and hospital- as well as community-associated antibiotic-resistant strains are an emerging global public health problem. Therefore, prophylactic vaccines or immune-based therapies are considered as alternative treatment opportunities. To develop such novel treatment approaches, a better understanding of the bacterial virulence and immune evasion mechanisms and their potential effects on immune-based therapies is essential. One important staphylococcal virulence factor is alpha-toxin, which is able to disrupt the epithelial barrier in order to establish infection. In addition, alpha-toxin has been reported to modulate other cell types including immune cells. Since CD4+ T cell-mediated immunity is required for protection against S. aureus infection, we were interested in the ability of alpha-toxin to directly modulate CD4+ T cells. To address this, murine naïve CD4+ T cells were differentiated in vitro into effector T cell subsets in the presence of alpha-toxin. Interestingly, alpha-toxin induced death of Th1-polarized cells, while cells polarized under Th17 conditions showed a high resistance toward increasing concentrations of this toxin. These effects could neither be explained by differential expression of the cellular alpha-toxin receptor ADAM10 nor by differential activation of caspases, but might result from an increased susceptibility of Th1 cells toward Ca2+-mediated activation-induced cell death. In accordance with the in vitro findings, an alpha-toxin-dependent decrease of Th1 and concomitant increase of Th17 cells was observed in vivo during S. aureus bacteremia. Interestingly, corresponding subsets of innate lymphoid cells and γδ T cells were similarly affected, suggesting a more general effect of alpha-toxin on the modulation of type 1 and type 3 immune responses. In conclusion, we have identified a novel alpha-toxin-dependent immunomodulatory strategy of S. aureus, which can directly act on CD4+ T cells and might be exploited for the development of novel immune-based therapeutic approaches to treat infections with antibiotic-resistant S. aureus strains.
Collapse
Affiliation(s)
- Agnes Bonifacius
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Oliver Goldmann
- Department Infection Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Floess
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Silva Holtfreter
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Philippe A Robert
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Maria Nordengrün
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Friederike Kruse
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Matthias Lochner
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research, Hanover, Germany.,Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hanover, Germany
| | - Christine S Falk
- Institute of Transplant Immunology, Hannover Medical School, Hanover, Germany.,DZIF, German Center for Infectious Diseases, TTU-IICH Hannover-Braunschweig Site, Hanover, Germany
| | - Ingo Schmitz
- Department Systems-Oriented Immunology and Inflammation Research, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Department of Molecular Immunology, Ruhr-University Bochum, Bochum, Germany
| | - Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Eva Medina
- Department Infection Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
40
|
Abstract
Osteomyelitis, or inflammation of bone, is most commonly caused by invasion of bacterial pathogens into the skeleton. Bacterial osteomyelitis is notoriously difficult to treat, in part because of the widespread antimicrobial resistance in the preeminent etiologic agent, the Gram-positive bacterium Staphylococcus aureus Bacterial osteomyelitis triggers pathological bone remodeling, which in turn leads to sequestration of infectious foci from innate immune effectors and systemically delivered antimicrobials. Treatment of osteomyelitis therefore typically consists of long courses of antibiotics in conjunction with surgical debridement of necrotic infected tissues. Even with these extreme measures, many patients go on to develop chronic infection or sustain disease comorbidities. A better mechanistic understanding of how bacteria invade, survive within, and trigger pathological remodeling of bone could therefore lead to new therapies aimed at prevention or treatment of osteomyelitis as well as amelioration of disease morbidity. In this minireview, we highlight recent developments in our understanding of how pathogens invade and survive within bone, how bacterial infection or resulting innate immune responses trigger changes in bone remodeling, and how model systems can be leveraged to identify new therapeutic targets. We review the current state of osteomyelitis epidemiology, diagnostics, and therapeutic guidelines to help direct future research in bacterial pathogenesis.
Collapse
|
41
|
Helbig L, Guehring T, Titze N, Nurjadi D, Sonntag R, Armbruster J, Wildemann B, Schmidmaier G, Gruetzner AP, Freischmidt H. A new sequential animal model for infection-related non-unions with segmental bone defect. BMC Musculoskelet Disord 2020; 21:329. [PMID: 32460740 PMCID: PMC7254709 DOI: 10.1186/s12891-020-03355-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The treatment of fracture-related infections (FRI) is still a challenge for orthopedic surgeons. The prevalence of FRI is particularly high in open fractures with extensive soft-tissue damage. This study aimed to develop a new two-step animal model for non-unions with segmental bone defects, which could be used to evaluate new innovative bone substitutes to improve the therapeutic options in humans with FRI and bone defects. METHODS After randomization to infected or non-infected groups, 30 Sprague-Dawley rats underwent a transverse osteotomy of the mid-shaft femur with a 5 mm defect. Additionally, the periosteum at the fracture zone was cauterized at both sides. After intramedullary inoculation with 103 CFU Staphylococcus aureus (infected group) or PBS (non-infected group), a fracture stabilization was done by intramedullary K-wires. After 5 weeks, the bone healing process was evaluated, and revision surgery was performed in order to obtain increased bone healing. The initial K-wires were removed, and debridement of the osteotomy-gap was done followed by a more stable re-osteosynthesis with an angle-stable plate. After further 8 weeks all rats were euthanized and the bone consolidation was tested biomechanically and the callus formation quantitatively by micro-CT analysis. RESULTS We developed and presented a new two-stage non-union animal model through a targeted S. aureus infection. After 5 weeks, all animals showed a non-union irrespective of assignment to the infected and non-infected group. Lane and Sandhu score showed a higher callus formation in the infected group. In all infected animals, the inoculated S. aureus strain was detected in the revision surgery. The second surgery did not improve bone healing, as shown by the Lane Sandhu score and in the μ-CT analysis. Similarly, biomechanical testing showed in both groups a significantly lower maximum torque as compared to the contralateral side (p < 0.0001). CONCLUSIONS We were able to successfully develop a new two-stage non-union animal model, which reflects a genuine clinical situation of an infection-related non-union model with segmental bone defects. This model could be used to evaluate various therapeutic anti-infectious and osteoinductive strategies in FRIs.
Collapse
Affiliation(s)
- Lars Helbig
- Clinic for Orthopedics and Trauma Surgery, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| | - Thorsten Guehring
- Arcus Sportklinik Pforzheim, Rastatterstr. 17-19, 75179, Pforzheim, Germany
| | - Nadine Titze
- Unfallklinik Ludwigshafen, Klinik für Unfallchirurgie und Orthopädie, Ludwig-Guttmann-Strasse 13, 67071, Ludwigshafen, Germany
| | - Dennis Nurjadi
- Department of Infectious Diseases Medical Microbiology and Hygiene, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Robert Sonntag
- Laboratory of Biomechanics and Implant Research, Clinic for Orthopedics and Trauma Surgery, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| | - Jonas Armbruster
- Unfallklinik Ludwigshafen, Klinik für Unfallchirurgie und Orthopädie, Ludwig-Guttmann-Strasse 13, 67071, Ludwigshafen, Germany
| | - Britt Wildemann
- Department of Trauma, Hand and Reconstructive Surgery, Experimental Trauma Surgery, Universitätsklinikum Jena, 07747, Jena, Germany.,Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353, Berlin, Germany
| | - Gerhard Schmidmaier
- Clinic for Orthopedics and Trauma Surgery, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| | - Alfred Paul Gruetzner
- Unfallklinik Ludwigshafen, Klinik für Unfallchirurgie und Orthopädie, Ludwig-Guttmann-Strasse 13, 67071, Ludwigshafen, Germany
| | - Holger Freischmidt
- Unfallklinik Ludwigshafen, Klinik für Unfallchirurgie und Orthopädie, Ludwig-Guttmann-Strasse 13, 67071, Ludwigshafen, Germany.
| |
Collapse
|
42
|
Hofstee MI, Muthukrishnan G, Atkins GJ, Riool M, Thompson K, Morgenstern M, Stoddart MJ, Richards RG, Zaat SAJ, Moriarty TF. Current Concepts of Osteomyelitis: From Pathologic Mechanisms to Advanced Research Methods. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1151-1163. [PMID: 32194053 DOI: 10.1016/j.ajpath.2020.02.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/18/2020] [Accepted: 02/27/2020] [Indexed: 01/18/2023]
Abstract
Osteomyelitis is an inflammation of the bone and bone marrow that is most commonly caused by a Staphylococcus aureus infection. Much of our understanding of the underlying pathophysiology of osteomyelitis, from the perspective of both host and pathogen, has been revised in recent years, with notable discoveries including the role played by osteocytes in the recruitment of immune cells, the invasion and persistence of S. aureus in submicron channels of cortical bone, and the diagnostic role of polymorphonuclear cells in implant-associated osteomyelitis. Advanced in vitro cell culture models, such as ex vivo culture models or organoids, have also been developed over the past decade, and have become widespread in many fields, including infectious diseases. These models better mimic the in vivo environment, allow the use of human cells, and can reduce our reliance on animals in osteomyelitis research. In this review, we provide an overview of the main pathologic concepts in osteomyelitis, with a focus on the new discoveries in recent years. Furthermore, we outline the value of modern in vitro cell culture techniques, with a focus on their current application to infectious diseases and osteomyelitis in particular.
Collapse
Affiliation(s)
- Marloes I Hofstee
- AO Research Institute Davos, Davos, Switzerland; Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research and Department of Orthopaedics, University of Rochester Medical Center, Rochester, New York
| | - Gerald J Atkins
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, South Australia, Australia
| | - Martijn Riool
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | | | - Mario Morgenstern
- Department of Orthopedic Surgery and Traumatology, University Hospital Basel, Basel, Switzerland
| | | | | | - Sebastian A J Zaat
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | | |
Collapse
|
43
|
Potential osteomyelitis biomarkers identified by plasma metabolome analysis in mice. Sci Rep 2020; 10:839. [PMID: 31964942 PMCID: PMC6972943 DOI: 10.1038/s41598-020-57619-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 01/02/2020] [Indexed: 01/18/2023] Open
Abstract
Osteomyelitis, which often arises from a surgical-site infection, is a serious problem in orthopaedic surgery. However, there are no specific biomarkers for osteomyelitis. Here, to identify specific plasma biomarkers for osteomyelitis, we conducted metabolome analyses using a mouse osteomyelitis model and bioluminescence imaging. We divided adult male pathogen-free BALB/C mice into control, sham-control, and infected groups. In the infected group, a bioluminescent Staphylococcus aureus strain was inoculated into the femur, and osteomyelitis was detected by bioluminescence imaging. We next analysed the metabolome, by comprehensively measuring all of the small molecules. This analysis identified 279 metabolites, 12 of which were significantly higher and 45 were significantly lower in the infected group than in the sham-control and control groups. Principal component analysis identified sphingosine as the highest loading factor. Several acyl carnitines and fatty acids, particularly ω-3 and ω-6 polyunsaturated fatty acids, were significantly lower in the infected group. Several metabolites in the tricarboxylic acid cycle were lower in the infected group than in the other groups. Thus, we identified two sphingolipids, sphinganine and sphingosine, as positive biomarkers for mouse osteomyelitis, and two components in the tricarboxylic acid cycle, two-oxoglutarate and succinic acid, as negative biomarkers.
Collapse
|
44
|
Krishnan AG, Biswas R, Menon D, Nair MB. Biodegradable nanocomposite fibrous scaffold mediated local delivery of vancomycin for the treatment of MRSA infected experimental osteomyelitis. Biomater Sci 2020; 8:2653-2665. [DOI: 10.1039/d0bm00140f] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The study shows the development of a biodegradable bi-functional composite scaffold that can reduce bacterial infection, while promotes bone regeneration in osteomyelitis, without the need for revision surgery.
Collapse
Affiliation(s)
- Amit G. Krishnan
- Amrita Centre for Nanosciences and Molecular Medicine
- Amrita Institute of Medical Sciences and Research Centre
- Amrita Vishwa Vidyapeetham
- India
| | - Raja Biswas
- Amrita Centre for Nanosciences and Molecular Medicine
- Amrita Institute of Medical Sciences and Research Centre
- Amrita Vishwa Vidyapeetham
- India
| | - Deepthy Menon
- Amrita Centre for Nanosciences and Molecular Medicine
- Amrita Institute of Medical Sciences and Research Centre
- Amrita Vishwa Vidyapeetham
- India
| | - Manitha B. Nair
- Amrita Centre for Nanosciences and Molecular Medicine
- Amrita Institute of Medical Sciences and Research Centre
- Amrita Vishwa Vidyapeetham
- India
| |
Collapse
|
45
|
Liposomal mupirocin holds promise for systemic treatment of invasive Staphylococcus aureus infections. J Control Release 2019; 316:292-301. [DOI: 10.1016/j.jconrel.2019.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 01/05/2023]
|
46
|
Lu X, Chen R, Lv J, Xu W, Chen H, Ma Z, Huang S, Li S, Liu H, Hu J, Nie L. High-resolution bimodal imaging and potent antibiotic/photodynamic synergistic therapy for osteomyelitis with a bacterial inflammation-specific versatile agent. Acta Biomater 2019; 99:363-372. [PMID: 31465882 DOI: 10.1016/j.actbio.2019.08.043] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/26/2019] [Accepted: 08/23/2019] [Indexed: 02/05/2023]
Abstract
Unsatisfactory diagnosis and therapy of osteomyelitis are still common but challenging issues for clinicians. To overcome these problems, a bacterial inflammation-specific multifunctional agent, denoted bovine serum albumin-manganese dioxide-ubiquicidin29-41-indocyanine green (ICG) -gentamicin (BMUIG), was synthesized for combined high-resolution bimodal imaging and antibiotic/photodynamic therapy for osteomyelitis. BMUIG binding affinity and antibacterial ability were assessed by using Staphylococcus aureus (S. aureus). Photoacoustic/magnetic resonance imaging was performed on a mouse model of acute osteomyelitis after intravenous injection of BMUIG. Then, myelitis-bearing mice were treated with antibiotic/photodynamic combination therapy. BMUIG specifically targeted S. aureus in comparison with non-targeted agents. In the osteomyelitis model, the infection area was identified accurately and quickly through ICG-based photoacoustic imaging and Mn2+-based T1 magnetic resonance imaging after injection of BMUIG. Furthermore, the manganese dioxide in BMUIG reacted with the locally produced hydrogen peroxide under acidic inflammatory conditions, continuously generating oxygen for enhanced photodynamic therapy. In combination with low-dose gentamicin, a synergistic antibacterial effect was observed and bone infection was resolved. In summary, a non-invasive accurate diagnosis and effective synergistic therapy for osteomyelitis was successfully developed using a bacterial inflammation-specific versatile agent, which would provide a sound theranostic strategy for common infectious diseases. STATEMENT OF SIGNIFICANCE: Osteomyelitis is one of the most serious consequences in orthopedics. However, its inaccurate diagnosis and low-effective antibiotic treatment are still common but challenging issues for clinicians. To overcome these problems, we uniquely designed a bacterial inflammation-specific multifunctional nanoagent, bovine serum albumin-manganese dioxide-ubiquicidin29-41-indocyanine green-gentamicin (BMUIG), for high-resolution bimodal imaging and antibiotic/photodynamic combined therapy of osteomyelitis. Herein, high-resolution imaging technologies refer to classic magnetic resonance imaging and emerging photoacoustic imaging. Photodynamic therapy is subtly introduced because of its safe and effective killing mechanism, which can synergize the bactericidal effect of antibiotics. As a result, we successfully realize non-invasive accurate diagnosis and effective synergistic therapy for osteomyelitis by virtue of the bacterial inflammation-specific versatile agent, which will serve as a promising candidate for sound theranostics in common infectious diseases.
Collapse
Affiliation(s)
- Xiaolin Lu
- Department of Orthopaedics, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, PR China
| | - Ronghe Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Jing Lv
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Weicai Xu
- Department of Orthopaedics, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, PR China
| | - Hongjiang Chen
- Department of Orthopaedics, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, PR China
| | - Zebin Ma
- Department of Orthopaedics, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, PR China
| | - Shanshan Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Shi Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China
| | - Heng Liu
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Jun Hu
- Department of Orthopaedics, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, PR China.
| | - Liming Nie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, PR China.
| |
Collapse
|
47
|
Abstract
The inflammation of bone tissue is called osteomyelitis, and most cases are caused by an infection with the bacterium Staphylococcus aureus. To date, the bone-building cells, osteoblasts, have been implicated in the progression of these infections, but not much is known about how the bone-resorbing cells, osteoclasts, participate. In this study, we show that S. aureus can infect osteoclasts and proliferate inside these cells, whereas bone-residing macrophages, immune cells related to osteoclasts, destroy the bacteria. These findings elucidate a unique role for osteoclasts to harbor bacteria during infection, providing a possible mechanism by which bacteria could evade destruction by the immune system. Osteomyelitis (OM), or inflammation of bone tissue, occurs most frequently as a result of bacterial infection and severely perturbs bone structure. OM is predominantly caused by Staphylococcus aureus, and even with proper treatment, OM has a high rate of recurrence and chronicity. While S. aureus has been shown to infect osteoblasts, it remains unclear whether osteoclasts (OCs) are also a target of intracellular infection. Here, we demonstrate the ability of S. aureus to intracellularly infect and divide within OCs. OCs were differentiated from bone marrow macrophages (BMMs) by exposure to receptor activator of nuclear factor kappa-B ligand (RANKL). By utilizing an intracellular survival assay and flow cytometry, we found that at 18 h postinfection the intracellular burden of S. aureus increased dramatically in cells with at least 2 days of RANKL exposure, while the bacterial burden decreased in BMMs. To further explore the signals downstream of RANKL, we manipulated factors controlling OC differentiation, NFATc1 and alternative NF-κB, and found that intracellular bacterial growth correlates with NFATc1 levels in RANKL-treated cells. Confocal and time-lapse microscopy in mature OCs showed a range of intracellular infection that correlated inversely with S. aureus-phagolysosome colocalization. The propensity of OCs to become infected, paired with their diminished bactericidal capacity compared to BMMs, could promote OM progression by allowing S. aureus to evade initial immune regulation and proliferate at the periphery of lesions where OCs are most abundant.
Collapse
|
48
|
G-CSF partially mediates bone loss induced by Staphylococcus aureus infection in mice. Clin Sci (Lond) 2019; 133:1297-1308. [PMID: 31175224 DOI: 10.1042/cs20181001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 05/28/2019] [Accepted: 06/07/2019] [Indexed: 12/13/2022]
Abstract
Bone loss in Staphylococcus aureus (S. aureus) osteomyelitis poses a serious challenge to orthopedic treatment. The present study aimed to elucidate how S. aureus infection in bone might induce bone loss. The C57BL/6 mice were injected with S. aureus (106 CFU/ml, 100 μl) or with the same amount of vehicle (control) via the tail vein. Microcomputed tomography (microCT) analysis showed bone loss progressing from week 1 to week 5 after infection, accompanied by a decreased number of osteocalcin-positive stained osteoblasts and the suppressed mRNA expression of Runx2 and osteocalcin. Transcriptome profiles of GSE30119 were downloaded and analyzed to determine the differences in expression of inflammatory factors between patients with S. aureus infected osteomyelitis and healthy controls, the data showed significantly higher mRNA expression of granulocyte colony-stimulating factor (G-CSF) in the whole blood from patients with S. aureus infection. Enzyme-linked immunosorbent assay (ELISA) analysis confirmed an increased level of G-CSF in the bone marrow and serum from S. aureus infected mice, which might have been due to the increased amount of F4/80+ macrophages. Interestingly, G-CSF neutralizing antibody treatment significantly rescued the bone loss after S. aureus infection, as evidenced by its roles in improving BV/TV and preserving osteocalcin- and osterix-positive stained cells. Importantly, we found that G-CSF level was significantly up-regulated in the serum from osteomyelitis patients infected by S. aureus Together, S. aureus infection might suppress the function of osteoblastic cells and induce progressive bone loss by up-regulating the level G-CSF, suggesting a therapeutic potential for G-CSF neutralization in combating bone loss in S. aureus osteomyelitis.
Collapse
|
49
|
Bedogni A, Bettini G, Bedogni G, Basso D, Gatti D, Valisena S, Brunello A, Sorio M, Berno T, Giannini S, Navaglia F, Plebani M, Nocini PF, Blandamura S, Saia G, Bertoldo F. Is vitamin D deficiency a risk factor for osteonecrosis of the jaw in patients with cancer? A matched case-control study. J Craniomaxillofac Surg 2019; 47:1203-1208. [PMID: 30929994 DOI: 10.1016/j.jcms.2019.03.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/11/2019] [Accepted: 03/06/2019] [Indexed: 01/07/2023] Open
Abstract
PURPOSE A previous case-control histomorphometric study showed higher odds of osteomalacia in patients with bisphosphonate-related osteonecrosis of the jaw (BRONJ). Vitamin D deficiency causes osteomalacia and may therefore be involved in the pathogenesis of BRONJ. The present case-control study aimed at testing such hypothesis. MATERIALS AND METHODS BRONJ+ and BRONJ- patients treated with bisphosphonates were matched by sex (same) and age (within 5 years). Serum 25-hydroxy-vitamin D (25-OH-D), parathyroid hormone, bone alkaline phosphatase, total procollagen type 1 amino-terminal propeptide, carboxy-terminal collagen crosslinks, Dickkopf WNT signaling pathway inhibitor 1 and sclerostin were measured. RESULTS The main outcome was vitamin D deficiency defined as 25-OH-D < 50 nmol/l. A total of 51 BRONJ+ and 73 BRONJ- patients were studied. The frequency (95% CI) of vitamin D deficiency was 59% (45%-72%) in BRONJ+ and 62% (48%-75%) in BRONJ- patients. This amounts to a difference of -3% (-22%-16%, p = 0.77) for BRONJ+ patients. Serum 25-hydroxy-vitamin D and parathyroid hormone were similar in BRONJ+ and BRONJ- patients. Among the bone metabolism markers, only sclerostin differed between the two groups, being higher in BRONJ+ patients. CONCLUSION The present matched case-control study suggests that vitamin D deficiency is not a risk factor for BRONJ.
Collapse
Affiliation(s)
- Alberto Bedogni
- Unit of Maxillofacial Surgery, Department of Neuroscience-DNS, University of Padua, Padua, Italy.
| | - Giordana Bettini
- Unit of Maxillofacial Surgery, Department of Neuroscience-DNS, University of Padua, Padua, Italy
| | - Giorgio Bedogni
- Clinical Epidemiology Unit, Liver Research Center, Basovizza, Trieste, Italy
| | - Daniela Basso
- Unit of Laboratory Medicine, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Davide Gatti
- Unit of Rheumatology, Department of Medicine, University of Verona, Verona, Italy
| | - Silvia Valisena
- Service of Traumatology, Orthopedics and Hand Surgery, Regional Hospital of Bellinzona, Switzerland
| | | | - Marco Sorio
- Unit of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Tamara Berno
- Hematology and Clinical Immunology Branch, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Sandro Giannini
- Unit of Internal Medicine, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Filippo Navaglia
- Unit of Laboratory Medicine, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Mario Plebani
- Unit of Laboratory Medicine, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Pier Francesco Nocini
- Unit of Oral and Maxillofacial Surgery, Department of Surgery, University of Verona, Verona, Italy
| | | | - Giorgia Saia
- Unit of Maxillofacial Surgery, Department of Neuroscience-DNS, University of Padua, Padua, Italy
| | - Francesco Bertoldo
- Unit of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
50
|
Tuchscherr L, Pöllath C, Siegmund A, Deinhardt-Emmer S, Hoerr V, Svensson CM, Thilo Figge M, Monecke S, Löffler B. Clinical S. aureus Isolates Vary in Their Virulence to Promote Adaptation to the Host. Toxins (Basel) 2019; 11:toxins11030135. [PMID: 30823631 PMCID: PMC6468552 DOI: 10.3390/toxins11030135] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/18/2019] [Accepted: 02/21/2019] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus colonizes epithelial surfaces, but it can also cause severe infections. The aim of this work was to investigate whether bacterial virulence correlates with defined types of tissue infections. For this, we collected 10–12 clinical S. aureus strains each from nasal colonization, and from patients with endoprosthesis infection, hematogenous osteomyelitis, and sepsis. All strains were characterized by genotypic analysis, and by the expression of virulence factors. The host–pathogen interaction was studied through several functional assays in osteoblast cultures. Additionally, selected strains were tested in a murine sepsis/osteomyelitis model. We did not find characteristic bacterial features for the defined infection types; rather, a wide range in all strain collections regarding cytotoxicity and invasiveness was observed. Interestingly, all strains were able to persist and to form small colony variants (SCVs). However, the low-cytotoxicity strains survived in higher numbers, and were less efficiently cleared by the host than the highly cytotoxic strains. In summary, our results indicate that not only destructive, but also low-cytotoxicity strains are able to induce infections. The low-cytotoxicity strains can successfully survive, and are less efficiently cleared from the host than the highly cytotoxic strains, which represent a source for chronic infections. The understanding of this interplay/evolution between the host and the pathogen during infection, with specific attention towards low-cytotoxicity isolates, will help to optimize treatment strategies for invasive and therapy-refractory infection courses.
Collapse
Affiliation(s)
- Lorena Tuchscherr
- Institute of Medical Microbiology, Jena University Hospital, 07747 Jena, Germany.
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, 07747 Jena, Germany.
| | - Christine Pöllath
- Institute of Medical Microbiology, Jena University Hospital, 07747 Jena, Germany.
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, 07747 Jena, Germany.
| | - Anke Siegmund
- Institute of Medical Microbiology, Jena University Hospital, 07747 Jena, Germany.
| | - Stefanie Deinhardt-Emmer
- Institute of Medical Microbiology, Jena University Hospital, 07747 Jena, Germany.
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, 07747 Jena, Germany.
| | - Verena Hoerr
- Institute of Medical Microbiology, Jena University Hospital, 07747 Jena, Germany.
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, 07747 Jena, Germany.
| | - Carl-Magnus Svensson
- Applied Systems Biology, Leibniz-Institute for Natural Product Research and Infection Biology, 07745 Jena, Germany.
| | - Marc Thilo Figge
- Applied Systems Biology, Leibniz-Institute for Natural Product Research and Infection Biology, 07745 Jena, Germany.
- Faculty of Biological Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany.
| | - Stefan Monecke
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany.
- Institute of Medical Microbiology and Hygiene, Medical Faculty Carl Gustav Carus, 01307 Dresden, Germany.
| | - Bettina Löffler
- Institute of Medical Microbiology, Jena University Hospital, 07747 Jena, Germany.
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, 07747 Jena, Germany.
| |
Collapse
|