1
|
Fearing BV, Romereim SM, Danelson K, Smykowski M, Barankevich M, Serbin R, Chintalapudi N, Davis J, Appt S, Burkart H, Seymour RB, Hsu JR. Development of a small animal bone-anchored limb replacement model for infection interventions. OTA Int 2025; 8:e366. [PMID: 40071169 PMCID: PMC11892714 DOI: 10.1097/oi9.0000000000000366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/11/2024] [Accepted: 12/08/2024] [Indexed: 03/14/2025]
Abstract
Purpose Osseointegration-associated infections are a critical barrier to widespread implementation of osseointegrated (OI) prosthetics. To address this challenge, a preclinical animal model must exist of the human model to test potential interventions. In this article, we describe a novel rabbit model of OI implant-related infection that can act as a platform for rapid translation and development of therapeutic approaches to combat these uniquely challenging infections. Methods A single-stage amputation was performed by exposure, transection, reaming, and tapping of the tibia, followed by placement of a 75-mm Ti-6Al-4V cortical screw implant. Muscle and skin were closed, and a prosthetic was attached to the screw. Hematology, clinical chemistry, and imaging were performed up to 8 weeks. High-resolution microCT and histology were conducted at terminal end points. Intraosseous vancomycin delivery was compared with intravenous delivery. Serum and bone marrow collection was conducted across a period of 5 hours. Results Rabbits maintained normal ambulation, mobility, diet, and weight throughout the study period. Clinical chemistry results indicate normal ranges over the study course. microCT and histology demonstrate osseointegration between the threads of the implant within the medullary cavity. Pharmacokinetic data determined that intraosseous vancomycin delivery results in significantly lower vancomycin concentrations systemically compared with intravenous delivery and higher peak vancomycin concentration within the tibial canal. Conclusion This preclinical translational model represents a reproducible small animal model of OI transtibial amputation that successfully recreates the bone-skin-implant interface, material-bone interactions to match human OI, and a similar immune response. Preclinical efficacy of infection interventions will be further explored with establishment of this model.
Collapse
Affiliation(s)
- Bailey V. Fearing
- Atrium Health—Carolinas Medical Center, Atrium Health Musculoskeletal Institute, Charlotte, NC
- Wake Forest University School of Medicine, Charlotte, NC
| | - Sarah M. Romereim
- Atrium Health—Carolinas Medical Center, Atrium Health Musculoskeletal Institute, Charlotte, NC
| | - Kerry Danelson
- Atrium Health Wake Forest Baptist, Wake Forest University School of Medicine, Orthopaedic Surgery and Rehabilitation, Winston-Salem, NC
| | - Matthew Smykowski
- Atrium Health—Carolinas Medical Center, Atrium Health Musculoskeletal Institute, Charlotte, NC
| | - Marina Barankevich
- Atrium Health—Carolinas Medical Center, Atrium Health Musculoskeletal Institute, Charlotte, NC
| | - Ryan Serbin
- Atrium Health—Carolinas Medical Center, Atrium Health Musculoskeletal Institute, Charlotte, NC
| | - Nainisha Chintalapudi
- Atrium Health—Carolinas Medical Center, Atrium Health Musculoskeletal Institute, Charlotte, NC
| | - Jana Davis
- PennState Health, Penn State Bone and Joint Institute, Hershey, PA
| | - Susan Appt
- Wake Forest University School of Medicine, Comparative Medicine, Winston-Salem, NC
| | - Heather Burkart
- Wake Forest University School of Medicine, Comparative Medicine, Winston-Salem, NC
| | - Rachel B. Seymour
- Atrium Health—Carolinas Medical Center, Atrium Health Musculoskeletal Institute, Charlotte, NC
- Wake Forest University School of Medicine, Charlotte, NC
| | - Joseph R. Hsu
- Atrium Health—Carolinas Medical Center, Atrium Health Musculoskeletal Institute, Charlotte, NC
- Wake Forest University School of Medicine, Charlotte, NC
| |
Collapse
|
2
|
Li W, Huang Y, Gu D, Peng S, Zhang B, Peng F, Zhang D, Li M, Xiao J, Jia Z, Qiu L. Ascorbate-loaded MgFe layered double hydroxide for osteomyelitis treatment. J Control Release 2025; 378:1045-1060. [PMID: 39740696 DOI: 10.1016/j.jconrel.2024.12.072] [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: 08/03/2024] [Revised: 12/15/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
Bacterial infections evoke considerable apprehension in orthopedics. Traditional antibiotic treatments exhibit cytotoxic effects and foster bacterial resistance, thereby presenting an ongoing and formidable obstacle in the realm of therapeutic interventions. Achieving bacterial eradication and osteogenesis are critical requirements for bone infection treatment. Herein, we design and fabricate a nanoenzyme-mimicking drug through the co-precipitation process, integrating MgFe layered double hydroxide with ascorbic acid (AA@LDH), to facilitate the simultaneous presence of these two unique functionalities. Within a bacterial acidic milieu, the degradation of the AA@LDH nanosystem prompts ascorbic acid to undergo a pro-oxidative transformation, generating an abundance of reactive oxygen species (ROS). These ROS overwhelm bacterial cellular processes, including nucleic acid replication, cell wall construction, virulence factor production, biosynthetic pathways, and energy generation. This disruption culminates in substantial bacterial mortality, as substantiated by RNA sequencing data. Hence, the AA@LDH nano system exhibits an in vitro antibacterial rate of approximately 100 % and 99 %, against S.aureus and E. coli, respectivaly. Additionally, the AA@LDH could directly accelerate osteogenic differentiation in vitro, evidenced by a 50 % increase in alkaline phosphatase activity and a 270 % improvement in extracellular matrix mineralization capability. Moreover, it enhances osteointegration process in vivo by favorably reshaping the osteogenic immune microenvironment. This innovative nanosystem for delivery offers new strategies that concurrently combat bacterial infections, mitigate inflammation, and induce tissue regeneration, marking a significant advancement in the realm of advanced materials and its applications.
Collapse
Affiliation(s)
- Wei Li
- Department of Sports Medicine, Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yuliang Huang
- Department of Traumatology and Orthopaedic Surgery, Huizhou Central People's Hospital, Huizhou 516001, China; Hui Zhou-Hong Kong Bone Health Joint Research Center, Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou 516001, China
| | - Dongqiang Gu
- Department of Sports Medicine, Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Sijun Peng
- Department of Traumatology and Orthopaedic Surgery, Huizhou Central People's Hospital, Huizhou 516001, China; Hui Zhou-Hong Kong Bone Health Joint Research Center, Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou 516001, China
| | - Bao Zhang
- Department of Orthopedics, Guangyuan Central Hospital, Guangyuan 628000, China
| | - Feng Peng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Dongdong Zhang
- Research Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mingjun Li
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China
| | - Jin Xiao
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| | - Zhiwei Jia
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 101100, China.
| | - Longhai Qiu
- Department of Traumatology and Orthopaedic Surgery, Huizhou Central People's Hospital, Huizhou 516001, China; Hui Zhou-Hong Kong Bone Health Joint Research Center, Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou 516001, China.
| |
Collapse
|
3
|
Liu X, Enoki Y, Taguchi K, Matsumoto K. Development of a pharmacokinetic/pharmacodynamic evaluation model for osteomyelitis and usefulness of tedizolid as an alternative to vancomycin against MRSA osteomyelitis. J Pharm Pharmacol 2025; 77:291-298. [PMID: 39504578 DOI: 10.1093/jpp/rgae124] [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] [Accepted: 10/17/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVES This study aimed to develop a suitable osteomyelitis model for pharmacokinetic/pharmacodynamic (PK/PD) evaluation and to investigate the target PK/PD values of vancomycin and tedizolid against methicillin-resistant Staphylococcus aureus (MRSA) osteomyelitis. METHODS An osteomyelitis model was established by implanting an MRSA-exposed sterilized suture in the tibia of normal mice and mice with cyclophosphamide-induced neutropenia. The suitability of the osteomyelitis mouse model for PK/PD evaluation was assessed using vancomycin as an indicator. The target PK/PD values for tedizolid were determined using this model. KEY FINDINGS In neutropenic mice, to achieve a static effect and 1 log10 kill against MRSA, the ratios of the area under the free drug concentration-time curve for 24 h to the minimum inhibitory concentration (fAUC24/MIC) of vancomycin were 91.29 and 430.03, respectively, confirming the validity of the osteomyelitis model for PK/PD evaluation. In immunocompetent mice, the target fAUC24/MIC values of tedizolid for achieving a static effect and 1 log10 kill against MRSA were 2.40 and 49.20, respectively. Additionally, only a 0.28 log10 kill was achieved in neutropenic mice with 20 times the human equivalent dose of tedizolid. CONCLUSIONS In patients with restored immunity, tedizolid can potentially be used as an alternative to intravenous vancomycin therapy.
Collapse
Affiliation(s)
- Xiaoxi Liu
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba koen, Minato-ku, Tokyo 105-8512, Japan
| | - Yuki Enoki
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba koen, Minato-ku, Tokyo 105-8512, Japan
| | - Kazuaki Taguchi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba koen, Minato-ku, Tokyo 105-8512, Japan
| | - Kazuaki Matsumoto
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba koen, Minato-ku, Tokyo 105-8512, Japan
| |
Collapse
|
4
|
Song L, Schwinn LS, Barthel J, Ketter V, Lechler P, Linne U, Rastan AJ, Vogt S, Ruchholtz S, Paletta JRJ, Günther M. Implant-Derived S. aureus Isolates Drive Strain-Specific Invasion Dynamics and Bioenergetic Alterations in Osteoblasts. Antibiotics (Basel) 2025; 14:119. [PMID: 40001363 PMCID: PMC11852183 DOI: 10.3390/antibiotics14020119] [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: 12/09/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Implants are integral to modern orthopedic surgery. The outcomes are good, but infections remain a serious issue. Staphylococcus aureus (S. aureus), along with Staphylococcus epidermidis, are predominant pathogens responsible for implant-associated infections, as conventional antibiotic treatments often fail due to biofilm formation or the pathogens' ability to invade cells and to persist intracellularly. Objectives: This study therefore focused on interactions of S. aureus isolates from infected implants with MG63 and SaOS2 osteoblasts by investigating the adhesion, invasion, and the impact on the bioenergetics of osteoblasts. Methods and Results: We found that the ability of S. aureus to adhere to osteoblasts depends on the isolate and was not associated with a single gene or expression pattern of characteristic adhesion proteins, and further, was not correlated with invasion. However, analysis of invasion capabilities identified better invasion conditions for S. aureus isolates with the SaOS2 osteoblastic cells. Interestingly, metabolic activity of osteoblasts remained unaffected by S. aureus infection, indicating cell survival. In contrast, respiration assays revealed an altered mitochondrial bioenergetic turnover in infected cells. While basal as well as maximal respiration in MG63 osteoblasts were not influenced statistically by S. aureus infections, we found increased non-mitochondrial respiration and enhanced glycolytic activity in the osteoblasts, which was again, more pronounced in the SaOS2 osteoblastic cells. Conclusions: Our findings highlight the complexity of S. aureus-host interactions, where both the pathogen and the host cell contribute to intracellular persistence and survival, representing a major factor for therapeutic failures.
Collapse
Affiliation(s)
- Lei Song
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Lea-Sophie Schwinn
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Juliane Barthel
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Vanessa Ketter
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Philipp Lechler
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Uwe Linne
- Faculty of Chemistry, Philipps-University Marburg, 35032 Marburg, Germany
| | - Ardawan J. Rastan
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Sebastian Vogt
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Steffen Ruchholtz
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Jürgen R. J. Paletta
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Madeline Günther
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| |
Collapse
|
5
|
Decodts M, Cantallops-Vilà C, Hornez JC, Lacroix JM, Bouchart F. Phage-Loaded Biomimetic Apatite Powder With Antibiofilm Activity to Treat Bone-Associated Infections. J Biomed Mater Res A 2025; 113:e37808. [PMID: 39376206 DOI: 10.1002/jbm.a.37808] [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: 06/12/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024]
Abstract
For decades, calcium phosphate (CaP)-based ceramics have been used for coating of bone and joint substitutes after arthroplasty due to their biocompatible properties. Infections following orthopedic replacement occur in 1%-5% of cases, causing serious complications. Biofilm formation either on the biomaterial's surface or on patient's tissues greatly enhances the resistance against antibiotic treatments and can induce a chronic infection, emphasizing the need for novel antimicrobial delivery systems. In this study, we established a protocol enabling bacteriophage loading during the synthesis of a CaP-based powder. The resulting biomaterial proved to be noncytotoxic against human osteoblastic cells and able to significantly inhibit 24-h matured S. aureus biofilm cultures or even completely eradicate it after 5 days of contact. Additional S. aureus biofilm assays with a freeze-dried material using two different excipients showed that sucrose had a protective role against Remus bacteriophage treatment of S. aureus biofilms, whereas lactose-freeze-dried powder maintained the antibiofilm activity.
Collapse
Affiliation(s)
- Maxime Decodts
- INSA Hauts-de-France, CERAMATHS-Laboratoire de Matériaux Céramiques et de Mathématiques, Univ. Polytechnique Hauts-de-France, Valenciennes, France
| | - Cristina Cantallops-Vilà
- INSA Hauts-de-France, CERAMATHS-Laboratoire de Matériaux Céramiques et de Mathématiques, Univ. Polytechnique Hauts-de-France, Valenciennes, France
| | - Jean-Christophe Hornez
- INSA Hauts-de-France, CERAMATHS-Laboratoire de Matériaux Céramiques et de Mathématiques, Univ. Polytechnique Hauts-de-France, Valenciennes, France
| | - Jean-Marie Lacroix
- UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Univ. Lille, CNRS, Lille, France
| | - Franck Bouchart
- INSA Hauts-de-France, CERAMATHS-Laboratoire de Matériaux Céramiques et de Mathématiques, Univ. Polytechnique Hauts-de-France, Valenciennes, France
- UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Univ. Lille, CNRS, Lille, France
| |
Collapse
|
6
|
Lee SH, Kim MB, Jeon YJ. Can bone SPECT/CT determine optimal sites for microbiological identification in post-traumatic or chronic osteomyelitis of extremities? Injury 2024; 55:111940. [PMID: 39426161 DOI: 10.1016/j.injury.2024.111940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024]
Abstract
INTRODUCTION Accurate microbiological identification is crucial when managing chronic osteomyelitis (COM) and post-traumatic osteomyelitis (PTO). Although bone single photon emission computed tomography/computed tomography (SPECT/CT) has helped in localizing osteomyelitis lesions, its effectiveness in guiding microbiological sampling remains unclear. This study aimed to determine whether bone SPECT/CT can improve microbiological identification rates in COM or PTO of the extremities. PATIENTS AND METHODS From February 2020 to August 2024, 53 patients with suspected COM or PTO in the extremities were retrospectively analyzed. All patients underwent bone SPECT/CT, followed by microbiological sampling during surgery. Tissue samples were taken from the areas of high SPECT/CT uptake or based on intraoperative findings where no uptake was observed. Microorganism identification rates were analyzed, including a sub-group analysis based on antibiotic discontinuation. RESULTS Of the 53 patients, 42 had positive bone SPECT/CT scan findings, with pathogen identification in 30 patients (71.4 %). In contrast, pathogen identification occurred in one out of twelve patients (9.1 %) with negative findings (odds ratio 25, p< 0.001). Bone SPECT/CT demonstrated a sensitivity of 96.8 % and an overall accuracy of 75.5 %. When antibiotics had been discontinued for ≥2 weeks, the pathogen identification rate increased to 90 %, compared with 50 % for <2 weeks of discontinuation (odds ratio 10.0, p= 0.006). In a sub-group of 30 patients with adequate antibiotic discontinuation duration, a positive bone SPECT/CT scan yielded a pathogen identification rate of 90.1 % (odds ratio 60.0, p= 0.001). CONCLUSION Bone SPECT/CT effectively identifies optimal sites for microbiological sampling in COM and PTO of the extremities, particularly when antibiotics have been discontinued for ≥2 weeks, enhancing pathogen detection rates.
Collapse
Affiliation(s)
- Seung Hoo Lee
- Department of Orthopaedic Surgery, Chungnam National University Sejong Hospital, Chungnam National University College of Medicine, Sejong, South Korea.
| | - Min Bom Kim
- Department of Orthopaedic Surgery, Seoul National University Hospital, Seoul National University College of Medicine, South Korea.
| | - Yeong June Jeon
- Department of Orthopaedic Surgery, Seoul National University Hospital, Seoul National University College of Medicine, South Korea.
| |
Collapse
|
7
|
Chang J, Kerr D, Zheng M, Seyler T. Chondrocyte Invasion May Be a Mechanism for Persistent Staphylococcus Aureus Infection In Vitro. Clin Orthop Relat Res 2024; 482:1839-1847. [PMID: 38662927 PMCID: PMC11419450 DOI: 10.1097/corr.0000000000003074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/11/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Recurrent bone and joint infection with Staphylococcus aureus is common. S. aureus can invade and persist in osteoblasts and fibroblasts, but little is known about this mechanism in chondrocytes. If S. aureus were able to invade and persist within chondrocytes, this could be a difficult compartment to treat. QUESTION/PURPOSE Can S. aureus infiltrate and persist intracellularly within chondrocytes in vitro? METHODS Cell lines were cultured in vitro and infected with S. aureus. Human chondrocytes (C20A4) were compared with positive controls of human osteoblasts (MG63) and mouse fibroblasts (NIH3T3), which have previously demonstrated S. aureus invasion and persistence (human fibroblasts were not available to us). Six replicates per cell type were followed for 6 days after infection. Cells were treated daily with antibiotic media for extracellular killing. To determine whether S. aureus can infiltrate chondrocytes, fluorescence microscopy was performed to qualitatively assess the presence of intracellular bacteria, and intracellular colony-forming units (CFU) were enumerated 2 hours after infection. To determine whether S. aureus can persist within chondrocytes, intracellular CFUs were enumerated from infected host cells each day postinfection. RESULTS S. aureus invaded human chondrocytes (C20A4) at a level (2.8 x 10 5 ± 5.5 x 10 4 CFUs/mL) greater than positive controls of human osteoblasts (MG63) (9.5 x 10 2 ± 2.5 x 10 2 CFUs/mL; p = 0.01) and mouse fibroblasts (NIH3T3) (9.1 x 10 4 ± 2.5 x 10 4 CFUs/mL; p = 0.02). S. aureus also persisted within human chondrocytes (C20A4) for 6 days at a level (1.4 x 10 3 ± 5.3 x 10 2 CFUs/mL) greater than that of human osteoblasts (MG63) (4.3 x 10 2 ± 3.5 x 10 1 CFUs/mL; p = 0.02) and mouse fibroblasts (NIH3T3) (0 CFUs/mL; p < 0.01). S. aureus was undetectable within mouse fibroblasts (NIH3T3) after 4 days. There were 0 CFUs yielded from cell media, confirming extracellular antibiotic treatment was effective. CONCLUSION S. aureus readily invaded human chondrocytes (C20A4) in vitro and persisted viably for 6 days after infection, evading extracellular antibiotics. Chondrocytes demonstrated a greater level of intracellular invasion and persistence by S. aureus than positive control human osteoblast (MG63) and mouse fibroblast (NIH3T3) cell lines. CLINICAL RELEVANCE Chondrocyte invasion and persistence may contribute to recurrent bone and joint infections. Additional research should assess longer periods of persistence and whether this mechanism is present in vivo.
Collapse
Affiliation(s)
- Jerry Chang
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - David Kerr
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Megan Zheng
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Thorsten Seyler
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| |
Collapse
|
8
|
Giordano V, Giannoudis PV. Biofilm Formation, Antibiotic Resistance, and Infection (BARI): The Triangle of Death. J Clin Med 2024; 13:5779. [PMID: 39407838 PMCID: PMC11476620 DOI: 10.3390/jcm13195779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Fracture-related infection (FRI) is a devastating event, directly affecting fracture healing, impairing patient function, prolonging treatment, and increasing healthcare costs. Time plays a decisive role in prognosis, as biofilm maturation leads to the development of antibiotic resistance, potentially contributing to infection chronicity and increasing morbidity and mortality. Research exploring the association between biofilm maturation and antibiotic resistance in orthopaedics primarily addresses aspects related to quality of life and physical function; however, little exists on life-threatening conditions and mortality. Understanding the intrinsic relationship between biofilm maturation, bacterial resistance, and mortality is critical in all fields of medicine. In the herein narrative review, we summarize recent evidence regarding biofilm formation, antibiotic resistance, and infection chronicity (BARI), the three basic components of the "triangle of death" of FRI, and its implications. Preoperative, perioperative, and postoperative prevention strategies to avoid the "triangle of death" of FRI are presented and discussed. Additionally, the importance of the orthopaedic trauma surgeon in understanding new tools to combat infections related to orthopaedic devices is highlighted.
Collapse
Affiliation(s)
- Vincenzo Giordano
- Serviço de Ortopedia e Traumatologia Prof. Nova Monteiro, Hospital Municipal Miguel Couto, Rua Mário Ribeiro 117/2º Andar, Gávea, Rio de Janeiro 22430-160, RJ, Brazil
| | - Peter V. Giannoudis
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds LS2 9LU, UK
- NIHR Leeds Biomedical Research Center, Chapel Allerton Hospital, Leeds LS7 4SA, UK
| |
Collapse
|
9
|
Taha M, AlDuwaisan A, Daneshmand M, Ibrahim MM, Bourget-Murray J, Grammatopoulos G, Garceau S, Abdelbary H. Mapping Staphylococcus aureus at Early and Late Stages of Infection in a Clinically Representative Hip Prosthetic Joint Infection Rat Model. Microorganisms 2024; 12:1895. [PMID: 39338569 PMCID: PMC11433939 DOI: 10.3390/microorganisms12091895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Prosthetic joint infection (PJI) continues to be a devastating complication following total joint replacement surgeries where Staphylococcus aureus is the main offending organism. To improve our understanding of the disease pathogenesis, a histological analysis of infected peri-implant tissue in a hip PJI rat model was utilized to assess S. aureus spread and tissue reaction at early and late stages of infection. Sprague-Dawley rats were used and received a left cemented hip hemiarthroplasty using a 3D-printed titanium femoral stem. The rats received an intra-articular injection of S. aureus Xen36. These infected rats were sacrificed either at 3 days post-infection (early-stage infection) or at 13-days post-infection (late-stage infection). The femoral and acetabular tissues of all animals were harvested at euthanasia. Histological analysis for the harvested tissue was performed using immunohistochemistry, hematoxylin and eosin, as well as Masson's trichrome stains. Histological examination revealed significant quantitative and qualitative differences in peri-implant tissue response to infection at early and late stages. This hip PJI rat model identified clear histologic differences between early and late stages of S. aureus infection and how quickly bacterial infiltration could occur. These findings can provide insight into why certain surgical strategies like debridement and antibiotics may be associated with high failure rates.
Collapse
Affiliation(s)
- Mariam Taha
- Chronic Disease Program, The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Division of Orthopaedic Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Abdullah AlDuwaisan
- Division of Orthopaedic Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
- Division of Orthopaedic Surgery, Faculty of Medicine, Kuwait University, Jabriya, Kuwait
| | - Manijeh Daneshmand
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mazen M Ibrahim
- Division of Orthopaedic Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | | | | | - Simon Garceau
- Division of Orthopaedic Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Hesham Abdelbary
- Chronic Disease Program, The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Division of Orthopaedic Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
10
|
Ledger EVK, Edwards AM. Host-induced cell wall remodeling impairs opsonophagocytosis of Staphylococcus aureus by neutrophils. mBio 2024; 15:e0164324. [PMID: 39041819 PMCID: PMC11323798 DOI: 10.1128/mbio.01643-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 07/24/2024] Open
Abstract
The bacterial pathogen Staphylococcus aureus responds to the host environment by increasing the thickness of its cell wall. However, the impact of cell wall thickening on susceptibility to host defenses is unclear. Using bacteria incubated in human serum, we show that host-induced increases in cell wall thickness led to a reduction in the exposure of bound antibody and complement and a corresponding reduction in phagocytosis and killing by neutrophils. The exposure of opsonins bound to protein antigens or lipoteichoic acid (LTA) was most significantly reduced, while opsonization by IgG against wall teichoic acid or peptidoglycan was largely unaffected. Partial digestion of accumulated cell wall using the enzyme lysostaphin restored opsonin exposure and promoted phagocytosis and killing. Concordantly, the antibiotic fosfomycin inhibited cell wall remodeling and maintained the full susceptibility of S. aureus to opsonophagocytic killing by neutrophils. These findings reveal that host-induced changes to the S. aureus cell wall reduce the ability of the immune system to detect and kill this pathogen through reduced exposure of protein- and LTA-bound opsonins. IMPORTANCE Understanding how bacteria adapt to the host environment is critical in determining fundamental mechanisms of immune evasion, pathogenesis, and the identification of targets for new therapeutic approaches. Previous work demonstrated that Staphylococcus aureus remodels its cell envelope in response to host factors and we hypothesized that this may affect recognition by antibodies and thus killing by immune cells. As expected, incubation of S. aureus in human serum resulted in rapid binding of antibodies. However, as bacteria adapted to the serum, the increase in cell wall thickness resulted in a significant reduction in exposure of bound antibodies. This reduced antibody exposure, in turn, led to reduced killing by human neutrophils. Importantly, while antibodies bound to some cell surface structures became obscured, this was not the case for those bound to wall teichoic acid, which may have important implications for vaccine design.
Collapse
Affiliation(s)
- Elizabeth V. K. Ledger
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Andrew M. Edwards
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| |
Collapse
|
11
|
Shi S, Lu W, Gu X, Lin Q. Efficacy of Gentamicin-Loaded Chitosan Nanoparticles Against Staphylococcus aureus Internalized in Osteoblasts. Microb Drug Resist 2024; 30:196-202. [PMID: 38579161 DOI: 10.1089/mdr.2023.0066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024] Open
Abstract
Staphylococcus aureus, the principal causative agent of osteomyelitis, can be internalized by osteoblasts and thereby escape from immune phagocytes and many kinds of antibiotics. To deliver antibiotics into osteoblasts to kill S. aureus in the intracellular environment, we developed gentamicin-loaded chitosan nanoparticles and evaluated their intracellular bactericidal effect. We found decreased numbers of S. aureus cells in infected osteoblasts treated with gentamicin-loaded chitosan nanoparticles. The cytotoxicity of the nanoparticles was evaluated by CCK-8 assay. There was no significant viability decrease at all tested concentrations. In conclusion, our results provide evidence for the potential use of gentamicin-loaded chitosan nanoparticles to enhance the delivery of gentamicin into cells and for their antibacterial effect against internalized S. aureus in the intracellular environment of osteoblasts.
Collapse
Affiliation(s)
- Sifeng Shi
- Department of Orthopedic Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wei Lu
- Department of Orthopedic Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xu Gu
- Department of Orthopedic Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qiang Lin
- Department of Orthopedic Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
12
|
Ferry T. A Review of Phage Therapy for Bone and Joint Infections. Methods Mol Biol 2024; 2734:207-235. [PMID: 38066372 DOI: 10.1007/978-1-0716-3523-0_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
There is a strong rationale for using phages in patients with bone and joint infections (BJIs). Indeed, specific phages can infect and replicate in bacterial pathogens and have also demonstrated their activity in vitro against biofilm produced by different bacteria. However, there is a high variability of the different clinical forms of BJI, and their management is complex and frequently includes surgery followed by the administration of antibiotics. Regardless of the availability of active phages, optimal ways of phage administration in patients with BJIs are unknown. Otherwise, all BJIs are not relevant for phage therapy. Except for diabetic foot infection, a BJI with bone exposure is potentially not a relevant indication for phage therapy. On the counterpart, prosthetic joint infections in patients for whom a multidisciplinary expert team judges a conservative approach as the best option to keep the patient's function seem to be a relevant indication with the hypothesis that phage therapy could increase the rate of infection control. The ESCMID Study Group for Non-traditional Antibacterial Therapy (ESGNTA) was created in 2022. One century after the first use of phages as a therapy, the phage therapy 2.0 era, with the possibility to evaluate personalized phage therapy in modern medicine and orthopedic surgery, is just open.
Collapse
Affiliation(s)
- Tristan Ferry
- Service de Maladies Infectieuses et Tropicales, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.
- Université Claude Bernard Lyon 1, Villeurbanne, France.
- Centre de Références des IOA Complexes de Lyon, CRIOAc Lyon, Lyon, France.
- StaPath team, Centre International de Recherche en Infectiologie, CIRI, Inserm U1111, CNRS UMR5308, ENS de Lyon, UCBL1, Lyon, France.
- Education and Clinical Officer of the ESCMID Study Group for Non-traditional Antibacterial Therapy (ESGNTA), Basel, Switzerland.
| |
Collapse
|
13
|
Kwon HK, Cahill SV, Yu KE, Alder KD, Dussik CM, Jeong J, Back JH, Lee FY. Parathyroid hormone therapy improves MRSA-infected fracture healing in a murine diabetic model. Front Cell Infect Microbiol 2023; 13:1230568. [PMID: 37829606 PMCID: PMC10565816 DOI: 10.3389/fcimb.2023.1230568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/28/2023] [Indexed: 10/14/2023] Open
Abstract
Introduction Diabetes mellitus (DM) impairs fracture healing and is associated with susceptibility to infection, which further inhibits fracture healing. While intermittent parathyroid hormone (1-34) (iPTH) effectively improves fracture healing, it is unknown whether infection-associated impaired fracture healing can be rescued with PTH (teriparatide). Methods A chronic diet-induced type 2 diabetic mouse model was used to yield mice with decreased glucose tolerance and increased blood glucose levels compared to lean-fed controls. Methicillin-resistant Staphylococcus aureus (MRSA) was inoculated in a surgical tibia fracture model to simulate infected fracture, after which mice were treated with a combination of antibiotics and adjunctive teriparatide treatment. Fracture healing was assessed by Radiographic Union Scale in Tibial Fractures (RUST), micro-computed tomography (μCT), biomechanical testing, and histology. Results RUST score was significantly poorer in diabetic mice compared to their lean nondiabetic counterparts. There were concomitant reductions in micro-computed tomography (μCT) parameters of callus architecture including bone volume/total volume, trabecular thickness, and total mineral density in type 2 diabetes mellitus (T2DM) mice. Biomechanicaltesting of fractured femora demonstrated diminished torsional rigidity, stiffness, and toughness to max torque. Adjuvant teriparatide treatment with systemic antibiotic therapy improved numerous parameters of bone microarchitecture bone volume, increased connectivity density, and increased trabecular number in both the lean and T2DM group. Despite the observation that poor fracture healing in T2DM mice was further impaired by MRSA infection, adjuvant iPTH treatment significantly improved fracture healing compared to antibiotic treatment alone in infected T2DM fractures. Discussion Our results suggest that teriparatide may constitute a viable adjuvant therapeutic agent to improve bony union and bone microarchitecture to prevent the development of septic nonunion under diabetic conditions.
Collapse
Affiliation(s)
- Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, United States
- Division of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Sean V. Cahill
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, United States
| | - Kristin E. Yu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, United States
| | - Kareme D. Alder
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, United States
| | - Christopher M. Dussik
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, United States
| | - Jain Jeong
- Department of Internal Medicine, Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, United States
| | - Jung Ho Back
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, United States
| | - Francis Y. Lee
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
14
|
Bhattacharjee B, Basak M, Das G, Ramesh A. Quinoxaline-based membrane-targeting therapeutic material: Implications in rejuvenating antibiotic and curb MRSA invasion in an in vitro bone cell infection model. BIOMATERIALS ADVANCES 2023; 148:213359. [PMID: 36963341 DOI: 10.1016/j.bioadv.2023.213359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/04/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
Manifestation of resistance in methicillin-resistant Staphylococcus aureus (MRSA) against multiple antibiotics demands an effective strategy to counter the menace of the pathogen. To address this challenge, the current study explores quinoxaline-based synthetic ligands as an adjuvant material to target MRSA in a combination therapy regimen. Amongst the tested ligands (C1-C4), only C2 was bactericidal against the MRSA strain S. aureus 4 s, with a minimum inhibitory concentration (MIC) of 32 μM. C2 displayed a membrane-directed activity and could effectively hinder MRSA biofilm formation. A quantitative real-time polymerase chain reaction (qRT-PCR) analysis indicated that C2 downregulated expression of the regulator gene agrC and reduced the fold change in the expression of adhesin genes fnbA and cnbA in MRSA in a dose-dependent manner. C2 enabled a 4-fold reduction in the MIC of ciprofloxacin (CPX) and in presence of 10 μM C2 and 8.0 μM CPX, growth of MRSA was arrested. Furthermore, a combination of 10 μM C2 and 12 μM CPX could strongly inhibit MRSA biofilm formation and reduce biofilm metabolic activity. The minimum biofilm inhibitory concentration (MBIC) of CPX against S. aureus 4 s biofilm was reduced and a synergy resulted between C2 and CPX. In a combinatorial treatment regimen, C2 could prevent emergence of CPX resistance and arrest growth of MRSA till 360 generations. C2 could also be leveraged in combination treatment (12 μM CPX and 10 μM C2) to target MRSA in an in vitro bone cell infection model, wherein MRSA cell adhesion and invasion onto cultured MG-63 cells was only ~17 % and ~ 0.37 %, respectively. The combinatorial treatment regimen was also biocompatible as the viability of MG-63 cells was high (~ 91 %). Thus, C2 is a promising adjuvant material to counter antibiotic-refractory therapy and mitigate MRSA-mediated bone cell infection.
Collapse
Affiliation(s)
- Basu Bhattacharjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Megha Basak
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Gopal Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Aiyagari Ramesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
15
|
Johnson MB, Furr KH, Suptela SR, Leach W, Marriott I. Induction of protective interferon-β responses in murine osteoblasts following Staphylococcus aureus infection. Front Microbiol 2022; 13:1066237. [PMID: 36532419 PMCID: PMC9757064 DOI: 10.3389/fmicb.2022.1066237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction The refractory and recurrent nature of chronic staphylococcal osteomyelitis may be due, at least in part, to the ability of Staphylococcus aureus to invade and persist within bone-forming osteoblasts. However, osteoblasts are now recognized to respond to S. aureus infection and produce numerous immune mediators and bone regulatory factors that can shape the host response. Type I interferons (IFNs) are best known for their antiviral effects, but it is becoming apparent that they impact host susceptibility to a wide range of pathogens including S. aureus. Methods Here, we have assessed the local expression of IFN-β by specific capture ELISA in an established in vivo mouse model of staphylococcal osteomyelitis. RNA Tag-Seq analysis, specific capture ELISAs, and/or immunoblot analyses, were then used to assess the expression of type I IFNs and select IFN stimulated genes (ISGs) in S. aureus infected primary murine osteoblasts. The effect of IFN-β on intracellular S. aureus burden was assessed in vitro following recombinant cytokine treatment by serial colony counts of liberated bacteria. Results We report the presence of markedly elevated IFN-β levels in infected bone tissue in a mouse model of staphylococcal osteomyelitis. RNA Tag-Seq analysis of S. aureus infected osteoblasts showed enrichment of genes associated with type I IFN signaling and ISGs, and elevated expression of mRNA encoding IFN-β and ISG products. IFN-β production was confirmed with the demonstration that S. aureus induces its rapid and robust release by osteoblasts in a dose-dependent manner. Furthermore, we showed increased protein expression of the ISG products IFIT1 and IFIT3 by infected osteoblasts and demonstrate that this occurs secondary to the release of IFN-β by these cells. Finally, we have determined that exposure of S. aureus-infected osteoblasts to IFN-β markedly reduces the number of viable bacteria harbored by these cells. Discussion Together, these findings indicate an ability of osteoblasts to respond to bacteria by producing IFN-β that can act in an autocrine and/or paracrine manner to elicit ISG expression and mitigate S. aureus infection.
Collapse
Affiliation(s)
- M. Brittany Johnson
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Kelli H. Furr
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Samantha R. Suptela
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Whitney Leach
- Department of Molecular Biology, Stowers Institute for Medical Research, Kansas City, MO, United States
| | - Ian Marriott
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| |
Collapse
|
16
|
Wang C, Lu H, Li X, Zhu Y, Ji Y, Lu W, Wang G, Dong W, Liu M, Wang X, Chen H, Tan C. Identification of an anti-virulence drug that reverses antibiotic resistance in multidrug resistant bacteria. Biomed Pharmacother 2022; 153:113334. [DOI: 10.1016/j.biopha.2022.113334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/14/2022] [Accepted: 06/23/2022] [Indexed: 11/02/2022] Open
|
17
|
Morita Y, Saito M, Rangel-Moreno J, Franchini AM, Owen JR, Martinez JC, Daiss JL, de Mesy Bentley KL, Kates SL, Schwarz EM, Muthukrishnan G. Systemic IL-27 administration prevents abscess formation and osteolysis via local neutrophil recruitment and activation. Bone Res 2022; 10:56. [PMID: 36028492 PMCID: PMC9418173 DOI: 10.1038/s41413-022-00228-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/20/2022] [Accepted: 06/15/2022] [Indexed: 01/07/2023] Open
Abstract
Interleukin-27 is a pleiotropic cytokine whose functions during bacterial infections remain controversial, and its role in patients with S. aureus osteomyelitis is unknown. To address this knowledge gap, we completed a clinical study and observed elevated serum IL-27 levels (20-fold higher, P < 0.05) in patients compared with healthy controls. Remarkably, IL-27 serum levels were 60-fold higher in patients immediately following septic death than in uninfected patients (P < 0.05), suggesting a pathogenic role of IL-27. To test this hypothesis, we evaluated S. aureus osteomyelitis in WT and IL-27Rα-/- mice with and without exogenous IL-27 induction by intramuscular injection of rAAV-IL-27p28 or rAAV-GFP, respectively. We found that IL-27 was induced at the surgical site within 1 day of S. aureus infection of bone and was expressed by M0, M1 and M2 macrophages and osteoblasts but not by osteoclasts. Unexpectedly, exogenous IL-27p28 (~2 ng·mL-1 in serum) delivery ameliorated soft tissue abscesses and peri-implant bone loss during infection, accompanied by enhanced local IL-27 expression, significant accumulation of RORγt+ neutrophils at the infection site, a decrease in RANK+ cells, and compromised osteoclast formation. These effects were not observed in IL-27Rα-/- mice compared with WT mice, suggesting that IL-27 is dispensable for immunity but mediates redundant immune and bone cell functions during infection. In vitro studies and bulk RNA-seq of infected tibiae showed that IL-27 increased nos1, nos2, il17a, il17f, and rorc expression but did not directly stimulate chemotaxis. Collectively, these results identify a novel phenomenon of IL-27 expression by osteoblasts immediately following S. aureus infection of bone and suggest a protective role of systemic IL-27 in osteomyelitis.
Collapse
Affiliation(s)
- Yugo Morita
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Motoo Saito
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Anthony M Franchini
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - John R Owen
- Department of Orthopedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - John C Martinez
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - John L Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Stephen L Kates
- Department of Orthopedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
18
|
Zelmer AR, Nelson R, Richter K, Atkins GJ. Can intracellular Staphylococcus aureus in osteomyelitis be treated using current antibiotics? A systematic review and narrative synthesis. Bone Res 2022; 10:53. [PMID: 35961964 PMCID: PMC9374758 DOI: 10.1038/s41413-022-00227-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/26/2022] [Accepted: 06/15/2022] [Indexed: 11/09/2022] Open
Abstract
Approximately 40% of treatments of chronic and recurrent osteomyelitis fail in part due to bacterial persistence. Staphylococcus aureus, the predominant pathogen in human osteomyelitis, is known to persist by phenotypic adaptation as small-colony variants (SCVs) and by formation of intracellular reservoirs, including those in major bone cell types, reducing susceptibility to antibiotics. Intracellular infections with S. aureus are difficult to treat; however, there are no evidence-based clinical guidelines addressing these infections in osteomyelitis. We conducted a systematic review of the literature to determine the demonstrated efficacy of all antibiotics against intracellular S. aureus relevant to osteomyelitis, including protein biosynthesis inhibitors (lincosamides, streptogramins, macrolides, oxazolidines, tetracyclines, fusidic acid, and aminoglycosides), enzyme inhibitors (fluoroquinolones and ansamycines), and cell wall inhibitors (beta-lactam inhibitors, glycopeptides, fosfomycin, and lipopeptides). The PubMed and Embase databases were screened for articles related to intracellular S. aureus infections that compared the effectiveness of multiple antibiotics or a single antibiotic together with another treatment, which resulted in 34 full-text articles fitting the inclusion criteria. The combined findings of these studies were largely inconclusive, most likely due to the plethora of methodologies utilized. Therefore, the reported findings in the context of the models employed and possible solutions for improved understanding are explored here. While rifampicin, oritavancin, linezolid, moxifloxacin and oxacillin were identified as the most effective potential intracellular treatments, the scientific evidence for these is still relatively weak. We advocate for more standardized research on determining the intracellular effectiveness of antibiotics in S. aureus osteomyelitis to improve treatments and patient outcomes.
Collapse
Affiliation(s)
- Anja R Zelmer
- Centre for Orthopaedic and Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Renjy Nelson
- Department of Infectious Diseases, Central Adelaide Local Health Network, Adelaide, SA, 5000, Australia.,Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Katharina Richter
- Richter Lab, Department of Surgery, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Adelaide, SA, 5011, Australia
| | - Gerald J Atkins
- Centre for Orthopaedic and Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5000, Australia.
| |
Collapse
|
19
|
Yu KE, Kwon HK, Dussik CM, Cahill SV, Back J, Alder KD, Lee FY. Enhancement of Impaired MRSA-Infected Fracture Healing by Combinatorial Antibiotics and Modulation of Sustained Inflammation. J Bone Miner Res 2022; 37:1352-1365. [PMID: 35616626 DOI: 10.1002/jbmr.4570] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/19/2022] [Accepted: 05/08/2022] [Indexed: 11/07/2022]
Abstract
Fracture healing is impaired in the setting of infection, which begets protracted inflammation. The most problematic causative agent of musculoskeletal infection is methicillin-resistant Staphylococcus aureus (MRSA). We hypothesized that modulation of excessive inflammation combined with cell-penetrating antibiotic treatments facilitates fracture healing in a murine MRSA-infected femoral fracture model. Sterile and MRSA-contaminated open transverse femoral osteotomies were induced in 10-week-old male C57BL/6 mice and fixed via intramedullary nailing. In the initial therapeutic cohort, empty, vancomycin (V), rifampin (R), vancomycin-rifampin (VR), or vancomycin-rifampin-trametinib (VRT) hydrogels were applied to the fracture site intraoperatively. Rifampin was included because of its ability to penetrate eukaryotic cells to target intracellular bacteria. Unbiased screening demonstrated ERK activation was upregulated in the setting of MRSA infection. As such, the FDA-approved mitogen-activated protein kinase kinase (MEK)1-pERK1/2 inhibitor trametinib was evaluated as an adjunctive therapeutic agent to selectively mitigate excessive inflammation after infected fracture. Two additional cohorts were created mimicking immediate and delayed postoperative antibiotic administration. Systemic vancomycin or VR was administered for 2 weeks, followed by 2 weeks of VRT hydrogel or oral trametinib therapy. Hematologic, histological, and cytokine analyses were performed using serum and tissue isolates obtained at distinct postoperative intervals. Radiography and micro-computed tomography (μCT) were employed to assess fracture healing. Pro-inflammatory cytokine levels remained elevated in MRSA-infected mice with antibiotic treatment alone, but increasingly normalized with trametinib therapy. Impaired callus formation and malunion were consistently observed in the MRSA-infected groups and was partially salvaged with systemic antibiotic treatment alone. Mice that received VR alongside adjuvant MEK1-pERK1/2 inhibition displayed the greatest restoration of bone and osseous union. A combinatorial approach involving adjuvant cell-penetrating antibiotic treatments alongside mitigation of excessive inflammation enhanced healing of infected fractures. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Kristin E Yu
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Christopher M Dussik
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Sean V Cahill
- Department of Orthopedic Surgery, Washington University, School of Medicine, St. Louis, MO, USA
| | - Jungho Back
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Kareme D Alder
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| |
Collapse
|
20
|
Alagboso FI, Mannala GK, Walter N, Docheva D, Brochhausen C, Alt V, Rupp M. Rifampicin restores extracellular organic matrix formation and mineralization of osteoblasts after intracellular Staphylococcus aureus infection. Bone Joint Res 2022; 11:327-341. [PMID: 35604422 PMCID: PMC9130678 DOI: 10.1302/2046-3758.115.bjr-2021-0395.r1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Aims Bone regeneration during treatment of staphylococcal bone infection is challenging due to the ability of Staphylococcus aureus to invade and persist within osteoblasts. Here, we sought to determine whether the metabolic and extracellular organic matrix formation and mineralization ability of S. aureus-infected human osteoblasts can be restored after rifampicin (RMP) therapy. Methods The human osteoblast-like Saos-2 cells infected with S. aureus EDCC 5055 strain and treated with 8 µg/ml RMP underwent osteogenic stimulation for up to 21 days. Test groups were Saos-2 cells + S. aureus and Saos-2 cells + S. aureus + 8 µg/ml RMP, and control groups were uninfected untreated Saos-2 cells and uninfected Saos-2 cells + 8 µg/ml RMP. Results The S. aureus-infected osteoblasts showed a significant number of intracellular bacteria colonies and an unusual higher metabolic activity (p < 0.005) compared to uninfected osteoblasts. Treatment with 8 µg/ml RMP significantly eradicated intracellular bacteria and the metabolic activity was comparable to uninfected groups. The RMP-treated infected osteoblasts revealed a significantly reduced amount of mineralized extracellular matrix (ECM) at seven days osteogenesis relative to uninfected untreated osteoblasts (p = 0.007). Prolonged osteogenesis and RMP treatment at 21 days significantly improved the ECM mineralization level. Ultrastructural images of the mineralized RMP-treated infected osteoblasts revealed viable osteoblasts and densely distributed calcium crystal deposits within the extracellular organic matrix. The expression levels of prominent bone formation genes were comparable to the RMP-treated uninfected osteoblasts. Conclusion Intracellular S. aureus infection impaired osteoblast metabolism and function. However, treatment with low dosage of RMP eradicated the intracellular S. aureus, enabling extracellular organic matrix formation and mineralization of osteoblasts at later stage. Cite this article: Bone Joint Res 2022;11(5):327–341.
Collapse
Affiliation(s)
- Francisca I Alagboso
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Gopala K Mannala
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Nike Walter
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany.,Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Denitsa Docheva
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany.,Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital Koenig-Ludwig-Haus, University of Wuerzburg, Wuerzburg, Germany
| | | | - Volker Alt
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany.,Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Markus Rupp
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany.,Department of Trauma Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Role of Implantable Drug Delivery Devices with Dual Platform Capabilities in the Prevention and Treatment of Bacterial Osteomyelitis. Bioengineering (Basel) 2022; 9:bioengineering9020065. [PMID: 35200418 PMCID: PMC8869141 DOI: 10.3390/bioengineering9020065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 11/26/2022] Open
Abstract
As medicine advances and physicians are able to provide patients with innovative solutions, including placement of temporary or permanent medical devices that drastically improve quality of life of the patient, there is the persistent, recurring problem of chronic bacterial infection, including osteomyelitis. Osteomyelitis can manifest as a result of traumatic or contaminated wounds or implant-associated infections. This bacterial infection can persist as a result of inadequate treatment regimens or the presence of biofilm on implanted medical devices. One strategy to mitigate these concerns is the use of implantable medical devices that simultaneously act as local drug delivery devices (DDDs). This classification of device has the potential to prevent or aid in clearing chronic bacterial infection by delivering effective doses of antibiotics to the area of interest and can be engineered to simultaneously aid in tissue regeneration. This review will provide a background on bacterial infection and current therapies as well as current and prospective implantable DDDs, with a particular emphasis on local DDDs to combat bacterial osteomyelitis.
Collapse
|
24
|
Chen K, Wang Y, Yuan Y, Qin W, Sheng YJ, Ahmed S, Sun C, Deng CL, Ojha SC. Molecular Tools for Guiding Therapy in Patients With Staphylococcal Bone and Joint Infections: A Diagnostic Test Accuracy Meta-analysis. Front Endocrinol (Lausanne) 2022; 13:792679. [PMID: 35909576 PMCID: PMC9326260 DOI: 10.3389/fendo.2022.792679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Timely detection of causative pathogens and their antimicrobial resistance are essential for guiding targeted therapies in bone and joint infections (BJI) patients. We performed a systematic review and meta-analysis to assess the diagnostic value of testing osteoarticular samples with the nucleic acid amplification tests (NAAT) for effective staphylococcal strain identification and the administration of appropriately targeted antimicrobial agents in BJI patients. METHODS Five databases, including PubMed, Embase, Scopus, Web of Science, and the Cochrane Library, were searched for related publications from inception to July 24, 2021. Studies comparing the diagnostic accuracy of NAAT to a microbiological culture reference standard of osteoarticular specimens were eligible. Pooled summary values of sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), and diagnostic odds ratio (DOR) of NAAT compared to the microbiological culture reference standard were calculated using bivariate random-effects meta-analyses. RESULTS From 906 citations, 11 studies were included. Eleven studies comprising 13 datasets (n = 1047) evaluated NAAT accuracy for methicillin-sensitive Staphylococcus aureus (MSSA) identification, while seven studies comprising nine datasets (n = 727) evaluated methicillin-resistant Staphylococcus aureus (MRSA) identification. Against the microbiological culture reference standard, the pooled summary estimates for detection of both MSSA [sensitivity: 0.89 (95% confidence interval [CI] 0.84-0.93), specificity: 0.99 (95% CI 0.97-0.99), PLR: 34.13 (95% CI 20.54-56.73), NLR: 0.19 (95% CI 0.12-0.3), and DOR: 283.37 (95% CI 129.49-620.1)] and MRSA [sensitivity: 0.81 (95% CI 0.67-0.91), specificity: 1.0 (95% CI 0.99-1.0), PLR: 62.1 (95% CI 24.5-157.6), NLR: 0.33 (95% CI 0.16-0.69), and DOR: 300.25 (95% CI 85.01-1060.5)] were comparable. Heterogeneity was moderate. GeneXpert was frequently used among NAA tests, and its diagnostic accuracy was in line with the overall pooled summary estimates. The heterogeneity in diagnostic efficacy (P >0.05) could not be explained by a meta-regression and subgroup analysis of the research design, sample condition, and patient selection technique. CONCLUSIONS Our study suggested that NAAT can be applied as the preferred prescreening test for the timely diagnosis of staphylococcal strains associated with BJI in osteoarticular samples for successful antimicrobial therapy.
Collapse
Affiliation(s)
- Ke Chen
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Southwest Medical University, Luzhou, China
| | - Yanqiu Wang
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yue Yuan
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wen Qin
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yun-Jian Sheng
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Southwest Medical University, Luzhou, China
| | - Sarfraz Ahmed
- Department of Basic Sciences, University of Veterinary and Animal Sciences Lahore, Narowal, Pakistan
| | - Changfeng Sun
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Southwest Medical University, Luzhou, China
| | - Cun-Liang Deng
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Suvash Chandra Ojha
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Southwest Medical University, Luzhou, China
- *Correspondence: Suvash Chandra Ojha,
| |
Collapse
|
25
|
Marro FC, Abad L, Blocker AJ, Laurent F, Josse J, Valour F. In vitro antibiotic activity against intraosteoblastic Staphylococcus aureus: a narrative review of the literature. J Antimicrob Chemother 2021; 76:3091-3102. [PMID: 34459881 PMCID: PMC8598303 DOI: 10.1093/jac/dkab301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Staphylococcus aureus – a major aetiological agent of bone and joint infection (BJI) – is associated with a high risk of relapse and chronicity, in part due to its ability to invade and persist in non-professional phagocytic bone cells such as osteoblasts. This intracellular reservoir protects S. aureus from the action of the immune system and most antibiotics. To date, the choice of antimicrobial strategies for BJI treatment mostly relies on standard susceptibility testing, bone penetration of antibiotics and their ‘antibiofilm’ activity. Despite the role of intracellular persistent S. aureus in the development of chronic infection, the ability of antibiotics to target the S. aureus intraosteoblastic reservoir is not considered in therapeutic choices but might represent a key determinant of treatment outcome. This review provides an overview of the intracellular pharmacokinetics of antistaphylococcal drugs used in the treatment of BJI and of their ability to target intraosteoblastic S. aureus. Thirteen studies focusing on the intraosteoblastic activity of antibiotics against S. aureus were reviewed, all relying on in vitro models of osteoblast infection. Despite varying incubation times, multiplicities of infection, bacterial strains, and the types of infected cell lines, rifamycins and fluoroquinolones remain the two most potent antimicrobial classes for intraosteoblastic S. aureus eradication, consistent with clinical data showing a superiority of this combination therapy in S. aureus orthopaedic device-related infections.
Collapse
Affiliation(s)
- Florian C Marro
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France.,Evotec ID Lyon, In Vitro Biology, Infectious Diseases and Antibacterials Unit, Gerland, 69007 Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France
| | - Lélia Abad
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
| | - Ariel J Blocker
- Evotec ID Lyon, In Vitro Biology, Infectious Diseases and Antibacterials Unit, Gerland, 69007 Lyon, France
| | - Frédéric Laurent
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France.,Centre de Référence pour la prise en charge des Infections ostéo-articulaires complexes (CRIOAc) Lyon, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Josse
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence pour la prise en charge des Infections ostéo-articulaires complexes (CRIOAc) Lyon, Hospices Civils de Lyon, Lyon, France
| | - Florent Valour
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence pour la prise en charge des Infections ostéo-articulaires complexes (CRIOAc) Lyon, Hospices Civils de Lyon, Lyon, France.,Service des maladies infectieuses et tropicales, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
26
|
Mukai M, Uchida K, Sugo K, Nakasu M, Nakajima T, Takata K, Takaso M, Urabe K. Long-term antibacterial activity of vancomycin from calcium phosphate cement in vivo. Biomed Mater Eng 2021; 33:41-50. [PMID: 34250926 DOI: 10.3233/bme-211243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Periprosthetic joint infection is a major complication of total joint arthroplasty, with treatment requiring a two-stage exchange procedure and 6 weeks of systemic antibiotics. However, depending on the infection site, intravenous delivery of antibiotics like vancomycin (VCM) can have poor tissue transferability, thus reducing their therapeutic effect. OBJECTIVE This study demonstrates the 24-week in vivo release profile and antibacterial activity of VCM from calcium phosphate cement impregnated with VCM (CPC/VCM) and compares them with those from polymethylmethacrylate impregnated with VCM (PMMA/VCM). METHODS Rats were implanted with the test specimens between the fascia and quadriceps. After implantation for 24 weeks, the test specimens were removed and residual VCM was extracted to calculate the concentration of VCM released into rat tissues. We also examined the antibacterial activity of releasable VCM from the removed test specimens by placing them directly onto the surface of agar. RESULTS CPC/VCM released greater concentrations of VCM for a longer period of time within the 24 weeks than PMMA/VCM. Moreover, CPC/VCM released 1.4 to 26.1-fold more VCM than PMMA/VCM. Using Staphylococcus aureus, antibacterial activity was logarithmically correlated with VCM concentration across the entire concentration range tested (12.5-800 μg/mL). While the area within which inhibition was observed-the inhibition zone-for both CPC/VCM and PMMA/VCM formed and gradually shrank with time after implantation, that for CPC/VCM was significantly larger than that for PMMA/VCM in each week after implantation. CONCLUSION CPC/VCM releases greater amounts of VCM with antibacterial activity for longer periods of time than PMMA/VCM, suggesting that CPC is effective for facilitating the release of antibiotics for local action in patients with established postoperative infection.
Collapse
Affiliation(s)
- Manabu Mukai
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Minami, Sagamihara, Kanagawa, Japan
| | - Kentaro Uchida
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Minami, Sagamihara, Kanagawa, Japan.,Shonan University of Medical Sciences Research Institute, Chigasaki, Kanagawa, Japan
| | - Ken Sugo
- Research and Development Department, HOYA Technosurgical Corporation, Akishima, Tokyo, Japan
| | - Masanori Nakasu
- Research and Development Department, HOYA Technosurgical Corporation, Akishima, Tokyo, Japan
| | - Takehiko Nakajima
- Research and Development Department, HOYA Technosurgical Corporation, Akishima, Tokyo, Japan
| | - Ken Takata
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Minami, Sagamihara, Kanagawa, Japan
| | - Masashi Takaso
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Minami, Sagamihara, Kanagawa, Japan
| | - Ken Urabe
- Department of Orthopaedic Surgery, Kitasato University Medical Center, Saitama, Japan
| |
Collapse
|
27
|
Schlatterer D. CORR Insights®: Tolerant Small-colony Variants Form Prior to Resistance Within a Staphylococcus aureus Biofilm Based on Antibiotic Selective Pressure. Clin Orthop Relat Res 2021; 479:1482-1483. [PMID: 33982975 PMCID: PMC8208395 DOI: 10.1097/corr.0000000000001790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 03/31/2021] [Indexed: 01/31/2023]
Affiliation(s)
- Daniel Schlatterer
- Vice Chair, Department of Orthopedic Surgery, Atlanta Medical Center, Atlanta, GA, USA
| |
Collapse
|
28
|
Kwon HK, Lee I, Yu KE, Cahill SV, Alder KD, Lee S, Dussik CM, Back J, Choi J, Song L, Kyriakides TR, Lee FY. Dual therapeutic targeting of intra-articular inflammation and intracellular bacteria enhances chondroprotection in septic arthritis. SCIENCE ADVANCES 2021; 7:eabf2665. [PMID: 34172438 PMCID: PMC8232912 DOI: 10.1126/sciadv.abf2665] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 05/12/2021] [Indexed: 05/10/2023]
Abstract
Bacterial infections involving joints and vital organs represent a challenging clinical problem because of the two concurrent therapeutic goals of bacterial eradication and tissue preservation. In the case of septic arthritis, permanent destruction of articular cartilage by intense host inflammation is commonly seen even after successful treatment of bacterial infection. Here, we provide scientific evidence of a novel treatment modality that can protect articular cartilage and enhanced eradication of causative bacteria in septic arthritis. Locally delivered cell-penetrating antibiotics such as rifampicin effectively eradicate intracellular reservoirs of methicillin-resistant Staphylococcus aureus within joint cells. Furthermore, mitigation of intra-articular inflammation by targeting the NLRP3 (nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing 3) inflammasome protects articular cartilage from damage in a murine model of knee septic arthritis. Together, concurrent mitigation of intra-articular inflammation and local adjuvant targeting of intracellular bacteria represents a promising new therapeutic strategy for septic arthritis.
Collapse
Affiliation(s)
- Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 800 Howard Ave., New Haven, CT 06510, USA
| | - Inkyu Lee
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 800 Howard Ave., New Haven, CT 06510, USA
| | - Kristin E Yu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 800 Howard Ave., New Haven, CT 06510, USA
| | - Sean V Cahill
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 800 Howard Ave., New Haven, CT 06510, USA
| | - Kareme D Alder
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 800 Howard Ave., New Haven, CT 06510, USA
| | - Saelim Lee
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 800 Howard Ave., New Haven, CT 06510, USA
| | - Christopher M Dussik
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 800 Howard Ave., New Haven, CT 06510, USA
| | - JungHo Back
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 800 Howard Ave., New Haven, CT 06510, USA
| | - Jeongjoon Choi
- Department of Microbial Pathogenesis, Yale School of Medicine, 295 Congress Ave., New Haven, CT 06536, USA
| | - Lee Song
- Department of Orthopedics Surgery, Columbia University, New York, NY 10032, USA
| | - Themis R Kyriakides
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, 800 Howard Ave., New Haven, CT 06510, USA.
| |
Collapse
|
29
|
Activity of Lysin CF-296 Alone and in Addition to Daptomycin in a Rat Model of Experimental Methicillin-Resistant Staphylococcus aureus Osteomyelitis. Antimicrob Agents Chemother 2021; 65:AAC.00117-21. [PMID: 33722889 DOI: 10.1128/aac.00117-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
30
|
Bose S, Sarkar N, Banerjee D. Natural medicine delivery from biomedical devices to treat bone disorders: A review. Acta Biomater 2021; 126:63-91. [PMID: 33657451 PMCID: PMC8247456 DOI: 10.1016/j.actbio.2021.02.034] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 12/28/2022]
Abstract
With an increasing life expectancy and aging population, orthopedic defects and bone graft surgeries are increasing in global prevalence. Research to date has advanced the understanding of bone biology and defect repair mechanism, leading to a marked success in the development of synthetic bone substitutes. Yet, the quest for functionalized bone grafts prompted the researchers to find a viable alternative that regulates cellular activity and supports bone regeneration and healing process without causing serious side-effects. Recently, researchers have introduced natural medicinal compounds (NMCs) in bone scaffold that enables them to release at a desirable rate, maintains a sustained release allowing sufficient time for tissue in-growth, and guides bone regeneration process with minimized risk of tissue toxicity. According to World Health Organization (WHO), NMCs are gaining popularity in western countries for the last two decades and are being used by 80% of the population worldwide. Compared to synthetic drugs, NMCs have a broader range of safety window and thus suitable for prolonged localized delivery for bone regeneration. There is limited literature focusing on the integration of bone grafts and natural medicines that provides detailed scientific evidences on NMCs, their toxic limits and particular application in bone tissue engineering, which could guide the researchers to develop functionalized implants for various bone disorders. This review will discuss the emerging trend of NMC delivery from bone grafts, including 3D-printed structures and surface-modified implants, highlighting the significance and potential of NMCs for bone health, guiding future paths toward the development of an ideal bone tissue engineering scaffold. STATEMENT OF SIGNIFICANCE: To date, additive manufacturing technology provids us with many advanced patient specific or defect specific bone constructs exhibiting three-dimensional, well-defined microstructure with interconnected porous networks for defect-repair applications. However, an ideal scaffold should also be able to supply biological signals that actively guide tissue regeneration while simultaneously preventing post-implantation complications. Natural biomolecules are gaining popularity in tissue engineering since they possess a safer, effective approach compared to synthetic drugs. The integration of bone scaffolds and natural biomolecules exploits the advantages of customized, multi-functional bone implants to provide localized delivery of biochemical signals in a controlled manner. This review presents an overview of bone scaffolds as delivery systems for natural biomolecules, which may provide prominent advancement in bone development and improve defect-healing caused by various musculoskeletal disorders.
Collapse
Affiliation(s)
- Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States.
| | - Naboneeta Sarkar
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States
| | - Dishary Banerjee
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States
| |
Collapse
|
31
|
Walsh L, Johnson CN, Hill C, Ross RP. Efficacy of Phage- and Bacteriocin-Based Therapies in Combatting Nosocomial MRSA Infections. Front Mol Biosci 2021; 8:654038. [PMID: 33996906 PMCID: PMC8116899 DOI: 10.3389/fmolb.2021.654038] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is a pathogen commonly found in nosocomial environments where infections can easily spread - especially given the reduced immune response of patients and large overlap between personnel in charge of their care. Although antibiotics are available to treat nosocomial infections, the increased occurrence of antibiotic resistance has rendered many treatments ineffective. Such is the case for methicillin resistant S. aureus (MRSA), which has continued to be a threat to public health since its emergence. For this reason, alternative treatment technologies utilizing antimicrobials such as bacteriocins, bacteriophages (phages) and phage endolysins are being developed. These antimicrobials provide an advantage over antibiotics in that many have narrow inhibition spectra, enabling treatments to be selected based on the target (pathogenic) bacterium while allowing for survival of commensal bacteria and thus avoiding collateral damage to the microbiome. Bacterial resistance to these treatments occurs less frequently than with antibiotics, particularly in circumstances where combinatory antimicrobial therapies are used. Phage therapy has been well established in Eastern Europe as an effective treatment against bacterial infections. While there are no Randomized Clinical Trials (RCTs) to our knowledge examining phage treatment of S. aureus infections that have completed all trial phases, numerous clinical trials are underway, and several commercial phage preparations are currently available to treat S. aureus infections. Bacteriocins have primarily been used in the food industry for bio-preservation applications. However, the idea of repurposing bacteriocins for human health is an attractive one considering their efficacy against many bacterial pathogens. There are concerns about the ability of bacteriocins to survive the gastrointestinal tract given their proteinaceous nature, however, this obstacle may be overcome by altering the administration route of the therapy through encapsulation, or by bioengineering protease-resistant variants. Obstacles such as enzymatic digestion are less of an issue for topical/local administration, for example, application to the surface of the skin. Bacteriocins have also shown impressive synergistic effects when used in conjunction with other antimicrobials, including antibiotics, which may allow antibiotic-based therapies to be used more sparingly with less resistance development. This review provides an updated account of known bacteriocins, phages and phage endolysins which have demonstrated an impressive ability to kill S. aureus strains. In particular, examples of antimicrobials with the ability to target MRSA strains and their subsequent use in a clinical setting are outlined.
Collapse
Affiliation(s)
- Lauren Walsh
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Crystal N Johnson
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Cork, Ireland
| | - Colin Hill
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R Paul Ross
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Cork, Ireland
| |
Collapse
|
32
|
Staphylococcus aureus cell wall structure and dynamics during host-pathogen interaction. PLoS Pathog 2021; 17:e1009468. [PMID: 33788901 PMCID: PMC8041196 DOI: 10.1371/journal.ppat.1009468] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/12/2021] [Accepted: 03/12/2021] [Indexed: 01/09/2023] Open
Abstract
Peptidoglycan is the major structural component of the Staphylococcus aureus cell wall, in which it maintains cellular integrity, is the interface with the host, and its synthesis is targeted by some of the most crucial antibiotics developed. Despite this importance, and the wealth of data from in vitro studies, we do not understand the structure and dynamics of peptidoglycan during infection. In this study we have developed methods to harvest bacteria from an active infection in order to purify cell walls for biochemical analysis ex vivo. Isolated ex vivo bacterial cells are smaller than those actively growing in vitro, with thickened cell walls and reduced peptidoglycan crosslinking, similar to that of stationary phase cells. These features suggested a role for specific peptidoglycan homeostatic mechanisms in disease. As S. aureus missing penicillin binding protein 4 (PBP4) has reduced peptidoglycan crosslinking in vitro its role during infection was established. Loss of PBP4 resulted in an increased recovery of S. aureus from the livers of infected mice, which coincided with enhanced fitness within murine and human macrophages. Thicker cell walls correlate with reduced activity of peptidoglycan hydrolases. S. aureus has a family of 4 putative glucosaminidases, that are collectively crucial for growth. Loss of the major enzyme SagB, led to attenuation during murine infection and reduced survival in human macrophages. However, loss of the other three enzymes Atl, SagA and ScaH resulted in clustering dependent attenuation, in a zebrafish embryo, but not a murine, model of infection. A combination of pbp4 and sagB deficiencies resulted in a restoration of parental virulence. Our results, demonstrate the importance of appropriate cell wall structure and dynamics during pathogenesis, providing new insight to the mechanisms of disease. The prevalence of methicillin resistant Staphylococcus aureus (MRSA) in both hospitals and the wider community places a huge weight on healthcare providers. To discover new control regimes, it is therefore important to understand how the pathogen behaves within the relevant environment of the host. This is often hampered by the ability to obtain sufficient ex vivo pathogen samples for study. We have developed a method to isolate S. aureus from the infected host to be able to analyse cellular morphology and structure. S. aureus, isolated from an infected kidney abscess are smaller in size, with thicker cell walls than exponentially growing cells in vitro. Their cell wall peptidoglycan also is less crosslinked. These features suggested the role of components controlling cell wall homeostasis as being important for infections. We tested the role of PBP4, known to increase cell wall crosslinking and found a pbp4 mutant to have increased survival in macrophages and fitness within the murine host. Conversely the peptidoglycan hydrolase SagB, whose loss results in thinner cell walls was attenuated in the murine systemic model of infection, with concomitant loss of fitness within macrophages. Our study reveals an important adaptation to the host environment and the role of those components involved in cell wall homeostasis in vivo.
Collapse
|
33
|
Staphylococcus aureus Internalization in Osteoblast Cells: Mechanisms, Interactions and Biochemical Processes. What Did We Learn from Experimental Models? Pathogens 2021; 10:pathogens10020239. [PMID: 33669789 PMCID: PMC7922271 DOI: 10.3390/pathogens10020239] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
Bacterial internalization is a strategy that non-intracellular microorganisms use to escape the host immune system and survive inside the human body. Among bacterial species, Staphylococcus aureus showed the ability to interact with and infect osteoblasts, causing osteomyelitis as well as bone and joint infection, while also becoming increasingly resistant to antibiotic therapy and a reservoir of bacteria that can make the infection difficult to cure. Despite being a serious issue in orthopedic surgery, little is known about the mechanisms that allow bacteria to enter and survive inside the osteoblasts, due to the lack of consistent experimental models. In this review, we describe the current knowledge about S. aureus internalization mechanisms and various aspects of the interaction between bacteria and osteoblasts (e.g., best experimental conditions, bacteria-induced damages and immune system response), focusing on studies performed using the MG-63 osteoblastic cell line, the best traditional (2D) model for the study of this phenomenon to date. At the same time, as it has been widely demonstrated that 2D culture systems are not completely indicative of the dynamic environment in vivo, and more recent 3D models—representative of bone infection—have also been investigated.
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
|
Alder KD, Lee I, Munger AM, Kwon HK, Morris MT, Cahill SV, Back J, Yu KE, Lee FY. Intracellular Staphylococcus aureus in bone and joint infections: A mechanism of disease recurrence, inflammation, and bone and cartilage destruction. Bone 2020; 141:115568. [PMID: 32745687 DOI: 10.1016/j.bone.2020.115568] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/19/2020] [Accepted: 07/26/2020] [Indexed: 02/06/2023]
Abstract
Bone and joint infections are devastating afflictions. Although medical interventions and advents have improved their care, bone and joint infections still portend dismal outcomes. Indeed, bone and joint infections are associated with extremely high mortality and morbidity rates and, generally, occur secondary to the aggressive pathogen Staphylococcus aureus. The consequences of bone and joint infections are further compounded by the fact that although they are aggressively treated, they frequently recur and result in massive bone and articular cartilage loss. Here, we review the literature and chronicle the fact that the fundamental cellular components of the musculoskeletal system can be internally infected with Staphylococcus aureus, which explains the ready recurrence of bone and joint infections even after extensive administration of antibiotic therapy and debridement and offer potential treatment solutions for further study. Moreover, we review the ramifications of intracellular infection and expound that the massive bone and articular cartilage loss is caused by the sustained proinflammatory state induced by infection and offer potential combination therapies for further study to protect bone and cartilage.
Collapse
Affiliation(s)
- Kareme D Alder
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Inkyu Lee
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Department of Life Science, Chung-Ang University, Seoul, Republic of Korea; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Alana M Munger
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Hyuk-Kwon Kwon
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Montana T Morris
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Sean V Cahill
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - JungHo Back
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Kristin E Yu
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| | - Francis Y Lee
- Department of Orthopædics & Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA; Yale University School of Medicine, Department of Orthopaedics and Rehabilitation, 330 Cedar St, TMP 523, PO Box 208071, New Haven, CT 06520-8071, USA.
| |
Collapse
|
36
|
Schmelcher M, Loessner MJ. Bacteriophage endolysins - extending their application to tissues and the bloodstream. Curr Opin Biotechnol 2020; 68:51-59. [PMID: 33126104 DOI: 10.1016/j.copbio.2020.09.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/25/2022]
Abstract
The rapid emergence of antibiotic-resistant bacteria and the lack of novel antibacterial agents pose a serious threat for patients and healthcare systems. Bacteriophage-encoded peptidoglycan hydrolases (endolysins) represent a promising new class of antimicrobials. Over the past two decades, research on these enzymes has evolved from basic in vitro characterization to sophisticated protein engineering approaches, including advanced preclinical and clinical testing. In recent years, increasingly specific animal models have shown efficacy of endolysins against bacterial infections of various different organs and tissues of the body. Despite these advances, some challenges with regard to systemic application of endolysins remain to be addressed. These include immunogenicity, circulation half-life, and cell and tissue-specific targeting and penetration properties.
Collapse
Affiliation(s)
- Mathias Schmelcher
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland.
| | - Martin J Loessner
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
Kamohara A, Hirata H, Xu X, Shiraki M, Yamada S, Zhang JQ, Kukita T, Toyonaga K, Hara H, Urano Y, Yamashita Y, Miyamoto H, Kukita A. IgG immune complexes with Staphylococcus aureus protein A enhance osteoclast differentiation and bone resorption by stimulating Fc receptors and TLR2. Int Immunol 2020; 32:89-104. [PMID: 31713625 DOI: 10.1093/intimm/dxz063] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 09/26/2019] [Indexed: 01/18/2023] Open
Abstract
Staphylococcus aureus is a main pathogen of osteomyelitis and protein A is a virulence factor with high affinity for IgG. In this study, we investigated whether S. aureus affects the differentiation and bone resorption of osteoclasts through the IgG-binding capacity of protein A. Staphylococcus aureus pre-treated with serum or IgG showed marked enhancement in osteoclastogenesis and bone resorption compared to non-treated S. aureus or a protein A-deficient mutant. Blocking of the Fc receptor and deletion of the Fcγ receptor gene in osteoclast precursor cells showed that enhanced osteoclastogenesis stimulated by S. aureus IgG immune complexes (ICs) was mediated by the Fc receptor on osteoclast precursor cells. In addition, osteoclastogenesis stimulated by S. aureus ICs but not the protein A-deficient mutant was markedly reduced in osteoclast precursor cells of Myd88-knockout mice. Moreover, NFATc1, Syk and NF-κB signals were necessary for osteoclastogenesis stimulated by S. aureus ICs. The results suggest the contribution of a of Toll-like receptor 2 (TLR2)-Myd88 signal to the activity of S. aureus ICs. We further examined the expression of pro-inflammatory cytokines that is known to be enhanced by FcγR-TLR cross-talk. Osteoclasts induced by S. aureus ICs showed higher expression of TNF-α and IL-1β, and marked stimulation of proton secretion of osteoclasts activated by pro-inflammatory cytokines. Finally, injection of S. aureus, but not the protein A-deficient mutant, exacerbated bone loss in implantation and intra-peritoneal administration mouse models. Our results provide a novel mechanistic aspect of bone loss induced by S. aureus in which ICs and both Fc receptors and TLR pathways are involved.
Collapse
Affiliation(s)
- Asana Kamohara
- Department of Pathology and Microbiology, Saga, Japan.,Department of Oral & Maxillofacial Surgery, Saga, Japan
| | - Hirohito Hirata
- Department of Pathology and Microbiology, Saga, Japan.,Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Xianghe Xu
- Department of Pathology and Microbiology, Saga, Japan.,Department of Molecular Cell Biology & Oral Anatomy, Faculty of Dentistry, Kyushu University, Fukuoka, Japan
| | - Makoto Shiraki
- Department of Pathology and Microbiology, Saga, Japan.,Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Sakuo Yamada
- Department of Medical Technology, Department of Clinical Nutrition, Faculty of Health Science & Technology, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
| | - Jing-Qi Zhang
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Toshio Kukita
- Department of Molecular Cell Biology & Oral Anatomy, Faculty of Dentistry, Kyushu University, Fukuoka, Japan
| | - Kenji Toyonaga
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Hiromitsu Hara
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Yasuteru Urano
- Department of Chemical Biology & Molecular Imaging, Graduate School of Medicine , Hongo, Tokyo, Japan.,Department of Chemistry & Biology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Tokyo, Japan
| | | | | | - Akiko Kukita
- Department of Pathology and Microbiology, Saga, Japan
| |
Collapse
|
38
|
Bibbo C, Mayer BE, Michetti LA. Foot and Ankle Surgery for Chronic Nonhealing Wounds. Surg Clin North Am 2020; 100:707-725. [DOI: 10.1016/j.suc.2020.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
39
|
Ferreira M, Aguiar S, Bettencourt A, Gaspar MM. Lipid-based nanosystems for targeting bone implant-associated infections: current approaches and future endeavors. Drug Deliv Transl Res 2020; 11:72-85. [PMID: 32514703 DOI: 10.1007/s13346-020-00791-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bone infections caused by Staphylococcus aureus are a major concern in medical care, particularly when associated with orthopedic-implant devices. The ability of the bacteria to form biofilms and their capacity to invade and persist within osteoblasts turn the infection eradication into a huge challenge. The reduction of antibiotic penetration through bacterial biofilms associated with the presence of persistent cells, ability to survive in the host, and high tolerance to antibiotics are some of the reasons for the difficult treatment of these infections. Effective therapeutic approaches are urgently needed. In this sense, lipid-based nanosystems, such as liposomes, have been investigated as an innovative and alternative strategy for the treatment of implant-associated S. aureus infections, due to their preferential accumulation at infected sites and interaction with S. aureus. This review highlights the recent advances on antibiotic-loaded liposome formulations both in vitro and in vivo and how the interaction with S. aureus biofilms may be improved by modulating the liposomal external surface. Graphical Abstract.
Collapse
Affiliation(s)
- Magda Ferreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
- Centre for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, Av. Universidade Técnica, 1300-477, Lisbon, Portugal
| | - Sandra Aguiar
- Centre for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, Av. Universidade Técnica, 1300-477, Lisbon, Portugal
| | - Ana Bettencourt
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
40
|
Watkins KE, Unnikrishnan M. Evasion of host defenses by intracellular Staphylococcus aureus. ADVANCES IN APPLIED MICROBIOLOGY 2020; 112:105-141. [PMID: 32762866 DOI: 10.1016/bs.aambs.2020.05.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Staphylococcus aureus is one of the leading causes of hospital and community-acquired infections worldwide. The increasing occurrence of antibiotic resistant strains and the high rates of recurrent staphylococcal infections have placed several treatment challenges on healthcare systems. In recent years, it has become evident that S. aureus is a facultative intracellular pathogen, able to invade and survive in a range of cell types. The ability to survive intracellularly provides this pathogen with yet another way to evade antibiotics and immune responses during infection. Intracellular S. aureus have been strongly linked to several recurrent infections, including severe bone infections and septicemias. S. aureus is armed with an array of virulence factors as well as an intricate network of regulators that enable it to survive, replicate and escape from a number of immune and nonimmune host cells. It is able to successfully manipulate host cell pathways and use it as a niche to multiply, disseminate, as well as persist during an infection. This bacterium is also known to adapt to the intracellular environment by forming small colony variants, which are metabolically inactive. In this review we will discuss the clinical evidence, the molecular pathways involved in S. aureus intracellular persistence, and new treatment strategies for targeting intracellular S. aureus.
Collapse
|
41
|
Abad L, Tafani V, Tasse J, Josse J, Chidiac C, Lustig S, Ferry T, Diot A, Laurent F, Valour F. Evaluation of the ability of linezolid and tedizolid to eradicate intraosteoblastic and biofilm-embedded Staphylococcus aureus in the bone and joint infection setting. J Antimicrob Chemother 2020; 74:625-632. [PMID: 30517641 DOI: 10.1093/jac/dky473] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/08/2018] [Accepted: 10/16/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES Prolonged use of linezolid for bone and joint infection (BJI) is limited by its long-term toxicity. The better safety profile of tedizolid, a recently developed oxazolidinone, could offer an alternative. However, its efficacy against biofilm-embedded and intracellular Staphylococcus aureus, the two main bacterial reservoirs associated with BJI chronicity, is unknown. METHODS Using three S. aureus strains (6850 and two clinical BJI isolates), linezolid and tedizolid were compared regarding their ability: (i) to target the S. aureus intracellular reservoir in an in vitro model of osteoblast infection, using three concentrations increasing from the bone concentration reached with standard therapeutic doses (Cbone = 2.5 × MIC; Cplasm = 10 × MIC; Cmax = 40 × MIC); (ii) to eradicate mature biofilm [minimal biofilm eradication concentration (MBEC)]; and (iii) to prevent biofilm formation [biofilm MIC (bMIC) and confocal microscopy]. RESULTS Linezolid and tedizolid weakly reduced the intracellular inoculum of S. aureus in a strain-dependent manner despite the similar MICs for the tested strains, but improved cell viability even in the absence of an intracellular bactericidal effect. Conversely, linezolid and tedizolid were ineffective in eradicating mature biofilm formed in vitro, with MBEC >2000 and >675 mg/L, respectively. bMICs of tedizolid were 4-fold lower than those of linezolid for all strains. CONCLUSIONS Linezolid and tedizolid alone are not optimal candidates to target bacterial phenotypes associated with chronic forms of BJI. Despite weak intracellular activity, they both reduce infection-related cytotoxicity, suggesting a role in modulating intracellular expression of staphylococcal virulence factors. Although inactive against biofilm-embedded S. aureus, both-but particularly tedizolid-are able to prevent biofilm formation.
Collapse
Affiliation(s)
- Lélia Abad
- CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France.,Regional Reference Centre for Complex Bone and Joint Infection (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Department of Bacteriology, Institute for Infectious Agents, Hospices Civils de Lyon, Lyon, France
| | - Virginie Tafani
- CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
| | - Jason Tasse
- CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
| | - Jérôme Josse
- CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
| | - Christian Chidiac
- CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France.,Regional Reference Centre for Complex Bone and Joint Infection (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Department of Infectious Diseases, Hospices Civils de Lyon, Lyon, France
| | - Sébastien Lustig
- Regional Reference Centre for Complex Bone and Joint Infection (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Department of Orthopaedic Surgery, Hospices Civils de Lyon, Lyon, France
| | - Tristan Ferry
- CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France.,Regional Reference Centre for Complex Bone and Joint Infection (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Department of Infectious Diseases, Hospices Civils de Lyon, Lyon, France
| | - Alan Diot
- CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
| | - Frédéric Laurent
- CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France.,Regional Reference Centre for Complex Bone and Joint Infection (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Department of Bacteriology, Institute for Infectious Agents, Hospices Civils de Lyon, Lyon, France
| | - Florent Valour
- CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France.,Regional Reference Centre for Complex Bone and Joint Infection (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Department of Infectious Diseases, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
42
|
Gerivani B, Staji H, Rassouli M, Ghazaleh N, Vayeghan AJ. Co-administration of Erythromycin and Leech Salivary Extract Alleviates Osteomyelitis in Rats Induced by Methicillin-Resistant Staphylococcus aureus. Vet Comp Orthop Traumatol 2020; 33:243-251. [PMID: 32356297 DOI: 10.1055/s-0040-1703008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Erythromycin (Ery) and leech saliva (LS) can inhibit Staphylococcus aureus growth in in vitro conditions. This study aimed to evaluate the activities and synergy between Ery and LS on chronic osteomyelitis in male Wistar rat's tibia induced by methicillin-resistant S. aureus (MRSA). MATERIALS AND METHODS Four weeks after osteomyelitis induction, rats were divided into four groups including no treatment (control), Ery monotherapy (orally), LS monotherapy, or Ery + LS twice daily for 2 weeks. Staphylococcus aureus growth, pathological signs and inflammatory cytokine tumour necrosis factor-alpha (TNF-α) levels were assessed. RESULTS Rats tolerated all therapeutic strategies well during the experiment. The Ery treatment alone significantly decreased bacterial growth, pathological signs and TNF-α levels. Leech saliva alone reduced TNF-α level significantly, but did not produce a significant reduction in bacterial growth and pathological signs. Ery + LS treatment significantly decreased bacterial growth, considerably alleviated bone pathological signs and decreased TNF-α levels compared with other groups. Statistical analysis suggested that there was a stronger efficiency and synergistic action of Ery and LS when combined against MRSA-induced osteomyelitis in rats. CLINICAL SIGNIFICANCE The present study suggests that LS may have clinical utility to treat MRSA-induced osteomyelitis when combined with Ery or other therapeutics.
Collapse
Affiliation(s)
- Bahar Gerivani
- Department of Pathobiology, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran (the Islamic Republic of)
| | - Hamid Staji
- Department of Pathobiology, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran (the Islamic Republic of)
| | - Maryam Rassouli
- Department of Pathobiology, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran (the Islamic Republic of)
| | - Nooshin Ghazaleh
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran (the Islamic Republic of)
| | - Abbas Javaheri Vayeghan
- Department of Pathobiology, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran (the Islamic Republic of)
| |
Collapse
|
43
|
Saunders RK, Infanti J, Ali H, Shuey T, Potteiger C, McNeilly S, Adams CS. Gram-Negative Bacteria Are Internalized Into Osteocyte-Like Cells. J Orthop Res 2020; 38:861-870. [PMID: 31692074 DOI: 10.1002/jor.24510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/14/2019] [Indexed: 02/04/2023]
Abstract
While Gram-positive organisms are the most common causative agent of initial bone infections, the percentage of Gram-negative species increases in reoccurring bone infections. As bacterial internalization has been suggested as one cause of reoccurring bone infection, we tested the hypothesis that Gram-negative species of bacteria can be internalized into bone cells. Using the MLO-A5 and the MLO-Y4 cell lines as our cell models, we demonstrated that the Gram-negative species, Proteus mirabilis and Serratia marcescens, can be internalized in these cells using an internalization assay. This rate at which these two species were internalized was both time- and initial concentration-dependent. Confocal analysis demonstrated the presence of internalized bacteria within both cell types. Inhibition of the cellular uptake with methyl-β-cyclodextrin and chloroquine both reduced internalized bacteria, indicating that this process is, at least in part, cell mediated. Finally, we demonstrated that the presence of internalized P. mirabilis did not impact cell viability, measured either by lactate dehydrogenase (LDH) release or 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) activity, while the presence of S. marcescens, on the other hand, both increased LDH release and reduced MTT activity, indicating a loss of cell viability in response to the organism. These results indicated that both species of Gram-negative bacteria can be internalized by bone cells and that these internalized bacteria could potentially result in reoccurring bone infections. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:861-870, 2020.
Collapse
Affiliation(s)
- Ray K Saunders
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joseph Infanti
- Graduate School of Biological Sciences, Rowan University, Stratford, New Jersey
| | - Hibah Ali
- Touro College of Osteopathic Medicine, New York, New York
| | - Timothy Shuey
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | - Courtney Potteiger
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | - Shelby McNeilly
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | - Christopher S Adams
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
44
|
Abad L, Josse J, Tasse J, Lustig S, Ferry T, Diot A, Laurent F, Valour F. Antibiofilm and intraosteoblastic activities of rifamycins against Staphylococcus aureus: promising in vitro profile of rifabutin. J Antimicrob Chemother 2020; 75:1466-1473. [DOI: 10.1093/jac/dkaa061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 12/14/2022] Open
Abstract
Abstract
Background
Targeting biofilm-embedded and intraosteoblastic Staphylococcus aureus, rifampicin gained a pivotal role in bone and joint infection (BJI) treatment. Two other rifamycins, rifabutin and rifapentine, may represent better-tolerated alternatives, but their activity against bacterial reservoirs associated with BJI chronicity has never been evaluated.
Objectives
To evaluate the activities of rifampicin, rifabutin and rifapentine in osteoblast infection models.
Methods
Using three S. aureus isolates, rifamycins were compared regarding: (i) their intracellular activity in ‘acute’ (24 h) and ‘chronic’ (7 days) osteoblast infection models at 0.1× MIC, 1× MIC, 10× MIC and 100× MIC, while impacting infection-induced cytotoxicity (MTT assay), intracellular phenol-soluble modulin (PSM) secretion (RT–PCR), resistance selection and small colony variant (SCV) emergence; and (ii) their minimal biofilm eradication concentration (MBEC) and their MIC to prevent biofilm formation (bMIC).
Results
At 0.1× MIC, only rifabutin significantly reduced intracellular inoculum and PSM secretion. All rifamycins allowed a 50% reduction of intraosteoblastic inoculum at higher concentrations, with no difference between acute and chronic infection models, while reducing infection-induced cytotoxicity and PSM secretion. Dose-dependent emergence of intracellular SCVs was observed for all molecules. No intracellular emergence of resistance was detected. bMICs were equivalent for all molecules, but MBEC90s of rifapentine and rifabutin were 10- to 100-fold lower than those of rifampicin, respectively.
Conclusions
All rifamycins are efficient in reducing the S. aureus intraosteoblastic reservoir while limiting infection-induced cytotoxicity, with a higher activity of rifabutin at low concentrations. All molecules prevent biofilm formation, but only rifapentine and rifabutin consistently reduce formed biofilm-embedded bacteria for all isolates. The activity of rifabutin at lower doses highlights its therapeutic potential.
Collapse
Affiliation(s)
- Lélia Abad
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
- Université Claude Bernard Lyon 1, Lyon, France
- Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Josse
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
| | - Jason Tasse
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
| | - Sébastien Lustig
- Université Claude Bernard Lyon 1, Lyon, France
- Centres de Référence pour la prise en charge des Infections ostéoarticulaires complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
- Department de chirurgie orthopédique, Hospices Civils de Lyon, Lyon, France
| | - Tristan Ferry
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
- Université Claude Bernard Lyon 1, Lyon, France
- Centres de Référence pour la prise en charge des Infections ostéoarticulaires complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
- Département maladies infectieuses et tropicales, Hospices Civils de Lyon, Lyon, France
| | - Alan Diot
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
| | - Frédéric Laurent
- Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
- Université Claude Bernard Lyon 1, Lyon, France
- Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
- Centres de Référence pour la prise en charge des Infections ostéoarticulaires complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
| | - Florent Valour
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
- Université Claude Bernard Lyon 1, Lyon, France
- Centres de Référence pour la prise en charge des Infections ostéoarticulaires complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
- Département maladies infectieuses et tropicales, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
45
|
Lüthje FL, Jensen LK, Jensen HE, Skovgaard K. The inflammatory response to bone infection - a review based on animal models and human patients. APMIS 2020; 128:275-286. [PMID: 31976582 DOI: 10.1111/apm.13027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/14/2020] [Indexed: 12/17/2022]
Abstract
Bone infections are difficult to diagnose and treat, especially when a prosthetic joint replacement or implant is involved. Bone loss is a major complication of osteomyelitis, but the mechanism behind has mainly been investigated in cell cultures and has not been confirmed in human settings. Inflammation is important in initiating an appropriate immune response to invading pathogens. However, many of the signaling molecules used by the immune system can also modulate bone remodeling and contribute to bone resorption during osteomyelitis. Our current knowledge of the inflammatory response relies heavily on animal models as research based on human samples is scarce. Staphylococcus aureus is one of the most common causes of bone infections and is the pathogen of choice in animal models. The regulation of inflammatory genes during prosthetic joint infections and implant-associated osteomyelitis has only been studied in rodent models. It is important to consider the validity of an animal model when results are extrapolated to humans, and both bone composition and the immune system of pigs has been shown to be more similar to humans, than to rodents. Here in vivo studies on the inflammatory response to prosthetic joint infections and implant-associated osteomyelitis are reviewed.
Collapse
Affiliation(s)
- Freja Lea Lüthje
- Department of Veterinary and Animal Science, University of Copenhagen, Frederiksberg C, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Louise Kruse Jensen
- Department of Veterinary and Animal Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Henrik Elvang Jensen
- Department of Veterinary and Animal Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
46
|
Yu K, Song L, Kang HP, Kwon HK, Back J, Lee FY. Recalcitrant methicillin-resistant Staphylococcus aureus infection of bone cells: Intracellular penetration and control strategies. Bone Joint Res 2020; 9:49-59. [PMID: 32435455 PMCID: PMC7229311 DOI: 10.1302/2046-3758.92.bjr-2019-0131.r1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
AIMS To characterize the intracellular penetration of osteoblasts and osteoclasts by methicillin-resistant Staphylococcus aureus (MRSA) and the antibiotic and detergent susceptibility of MRSA in bone. METHODS Time-lapse confocal microscopy was used to analyze the interaction of MRSA strain USA300 with primary murine osteoblasts and osteoclasts. The effects of early and delayed antibiotic treatments on intracellular and extracellular bacterial colony formation and cell death were quantified. We tested the effects of cefazolin, gentamicin, vancomycin, tetracycline, rifampicin, and ampicillin, as well as agents used in surgical preparation and irrigation. RESULTS MRSA infiltrated bone-resident cells within 15 to 30 minutes. Penetration was most effectively prevented with early (i.e. 30 minutes) antibiotic administration. The combined administration of rifampicin with other antibiotics potentiated their protective effects against MRSA-induced cytotoxicity and most significantly reduced extracellular bacterial bioburden. Gentamicin-containing compounds were most effective in reducing intracellular MRSA bioburden. Of the surgical preparation agents evaluated, betadine reduced in vitro MRSA growth to the greatest extent. CONCLUSION The standard of care for open fractures involves debridement and antibiotics within the first six hours of injury but does not account for the window in which bacteria penetrate cells. Antibiotics must be administered as early as possible after injury or prior to incision to prevent intracellular infestation. Rifampicin can potentiate the capacity of antibiotic regimens to reduce MRSA-induced cytotoxicity.Cite this article: Bone Joint Res. 2020;9(2):49-59.
Collapse
Affiliation(s)
- Kristin Yu
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| | - Lee Song
- Department of Orthopaedics, Columbia University, New York, New York, USA
| | - Hyunwoo Paco Kang
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| | - Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| | - Jungho Back
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
47
|
Dubois J, Dubois M. Levonadifloxacin (WCK 771) exerts potent intracellular activity against Staphylococcus aureus in THP-1 monocytes at clinically relevant concentrations. J Med Microbiol 2019; 68:1716-1722. [DOI: 10.1099/jmm.0.001102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
48
|
Labruère R, Sona AJ, Turos E. Anti-Methicillin-Resistant Staphylococcus aureus Nanoantibiotics. Front Pharmacol 2019; 10:1121. [PMID: 31636560 PMCID: PMC6787278 DOI: 10.3389/fphar.2019.01121] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/30/2019] [Indexed: 01/08/2023] Open
Abstract
Nanoparticle-based antibiotic constructs have become a popular area of investigation in the biomedical sciences. Much of this work has pertained to human diseases, largely in the cancer therapy arena. However, considerable research has also been devoted to the nanochemistry for controlling infectious diseases. Among these are ones due to bacterial infections, which can cause serious illnesses leading to death. The onset of multi-drug-resistant (MDR) infections such as those caused by the human pathogen Staphylococcus aureus has created a dearth of problems such as surgical complications, persistent infections, and lack of available treatments. In this article, we set out to review the primary literature on the design and development of new nanoparticle materials for the potential treatment of S. aureus infections, and areas that could be further expanded upon to make nanoparticle antibiotics a mainstay in clinical settings.
Collapse
Affiliation(s)
- Raphaël Labruère
- Institut de Chimie Moléculaire et des Matériaux d’Orsay (ICMMO), CNRS, Univ Paris Sud, Université Paris-Saclay, Orsay, France
| | - A. J. Sona
- Center for Molecular Diversity in Drug Design, Discovery and Delivery, Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Edward Turos
- Center for Molecular Diversity in Drug Design, Discovery and Delivery, Department of Chemistry, University of South Florida, Tampa, FL, United States
| |
Collapse
|
49
|
Masters EA, Trombetta RP, de Mesy Bentley KL, Boyce BF, Gill AL, Gill SR, Nishitani K, Ishikawa M, Morita Y, Ito H, Bello-Irizarry SN, Ninomiya M, Brodell JD, Lee CC, Hao SP, Oh I, Xie C, Awad HA, Daiss JL, Owen JR, Kates SL, Schwarz EM, Muthukrishnan G. Evolving concepts in bone infection: redefining "biofilm", "acute vs. chronic osteomyelitis", "the immune proteome" and "local antibiotic therapy". Bone Res 2019; 7:20. [PMID: 31646012 PMCID: PMC6804538 DOI: 10.1038/s41413-019-0061-z] [Citation(s) in RCA: 320] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/17/2019] [Accepted: 06/21/2019] [Indexed: 02/08/2023] Open
Abstract
Osteomyelitis is a devastating disease caused by microbial infection of bone. While the frequency of infection following elective orthopedic surgery is low, rates of reinfection are disturbingly high. Staphylococcus aureus is responsible for the majority of chronic osteomyelitis cases and is often considered to be incurable due to bacterial persistence deep within bone. Unfortunately, there is no consensus on clinical classifications of osteomyelitis and the ensuing treatment algorithm. Given the high patient morbidity, mortality, and economic burden caused by osteomyelitis, it is important to elucidate mechanisms of bone infection to inform novel strategies for prevention and curative treatment. Recent discoveries in this field have identified three distinct reservoirs of bacterial biofilm including: Staphylococcal abscess communities in the local soft tissue and bone marrow, glycocalyx formation on implant hardware and necrotic tissue, and colonization of the osteocyte-lacuno canalicular network (OLCN) of cortical bone. In contrast, S. aureus intracellular persistence in bone cells has not been substantiated in vivo, which challenges this mode of chronic osteomyelitis. There have also been major advances in our understanding of the immune proteome against S. aureus, from clinical studies of serum antibodies and media enriched for newly synthesized antibodies (MENSA), which may provide new opportunities for osteomyelitis diagnosis, prognosis, and vaccine development. Finally, novel therapies such as antimicrobial implant coatings and antibiotic impregnated 3D-printed scaffolds represent promising strategies for preventing and managing this devastating disease. Here, we review these recent advances and highlight translational opportunities towards a cure.
Collapse
Affiliation(s)
- Elysia A. Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY USA
| | - Ryan P. Trombetta
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY USA
| | - Karen L. de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY USA
| | - Brendan F Boyce
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY USA
| | - Ann Lindley Gill
- Department of Microbiology & Immunology, University of Rochester Medical Center, Rochester, NY USA
| | - Steven R. Gill
- Department of Microbiology & Immunology, University of Rochester Medical Center, Rochester, NY USA
| | - Kohei Nishitani
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
| | - Masahiro Ishikawa
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
| | - Yugo Morita
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
| | - Hiromu Ito
- Department of Orthopaedic Surgery, Kyoto University, Kyoto, Japan
| | | | - Mark Ninomiya
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
| | - James D. Brodell
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
| | - Charles C. Lee
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
| | - Stephanie P. Hao
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
| | - Irvin Oh
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY USA
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY USA
| | - Hani A. Awad
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY USA
| | - John L. Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY USA
| | - John R. Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, VA USA
| | - Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, VA USA
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY USA
- Department of Microbiology & Immunology, University of Rochester Medical Center, Rochester, NY USA
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY USA
| |
Collapse
|
50
|
Masters EA, Salminen AT, Begolo S, Luke EN, Barrett SC, Overby CT, Gill AL, de Mesy Bentley KL, Awad HA, Gill SR, Schwarz EM, McGrath JL. An in vitro platform for elucidating the molecular genetics of S. aureus invasion of the osteocyte lacuno-canalicular network during chronic osteomyelitis. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 21:102039. [PMID: 31247310 DOI: 10.1016/j.nano.2019.102039] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 11/30/2022]
Abstract
Staphylococcus aureus osteomyelitis is a devasting disease that often leads to amputation. Recent findings have shown that S. aureus is capable of invading the osteocyte lacuno-canalicular network (OLCN) of cortical bone during chronic osteomyelitis. Normally a 1 μm non-motile cocci, S. aureus deforms smaller than 0.5 μm in the sub-micron channels of the OLCN. Here we present the μSiM-CA (Microfluidic - Silicon Membrane - Canalicular Array) as an in vitro screening platform for the genetic mechanisms of S. aureus invasion. The μSiM-CA platform features an ultrathin silicon membrane with defined pores that mimic the openings of canaliculi. While we anticipated that S. aureus lacking the accessory gene regulator (agr) quorum-sensing system would not be capable of invading the OLCN, we found no differences in propagation compared to wild type in the μSiM-CA. However the μSiM-CA proved predictive as we also found that the agr mutant strain invaded the OLCN of murine tibiae.
Collapse
Affiliation(s)
- Elysia A Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY; Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY
| | - Alec T Salminen
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY
| | | | - Emma N Luke
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY
| | - Sydney C Barrett
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY
| | - Clyde T Overby
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY; Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY
| | - Ann Lindley Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY; Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY; Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY
| | - Hani A Awad
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY; Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY
| | - Steven R Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY; Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY; Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY; Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY.
| |
Collapse
|