1
|
Collet Q, Velard F, Laurent F, Josse J. Intracellular Staphylococcus aureus in osteoblasts and osteocytes and its impact on bone homeostasis during osteomyelitis. Bone 2025; 198:117536. [PMID: 40393553 DOI: 10.1016/j.bone.2025.117536] [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: 03/24/2025] [Revised: 05/13/2025] [Accepted: 05/16/2025] [Indexed: 05/22/2025]
Abstract
Osteomyelitis is a severe infection of bone tissue that can lead to bone loss and even osteonecrosis. This condition is mostly caused by Gram-positive bacteria, with Staphylococcus aureus being the most common etiological agent. Among the pathophysiological mechanisms involved in osteomyelitis, the ability of S. aureus to be internalized by osteoblasts or osteocytes and to survive within these cells, is particularly noteworthy. Infected osteoblasts and osteocytes not only serve as reservoirs in chronic cases of osteomyelitis but also play an active role in the osteoimmunology process, notably by producing mediators that promote the bone resorption activity of osteoclasts, thereby disrupting bone homeostasis. The present review explores both historical and recent literature on the internalization of S. aureus by osteoblasts and osteocytes, its intracellular behavior following internalization, and its mechanisms for inducing cell death. Additionally, it examines how S. aureus affects bone formation activity and promotes the production of inflammatory and pro-osteoclastic mediators. This review aims to highlight the limitations of current findings and outline key questions for future investigations.
Collapse
Affiliation(s)
- Quentin Collet
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, Lyon, France; Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de 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, Univ Lyon, 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, Lyon, France
| |
Collapse
|
2
|
Consoli FMA, Reyes-Matute A, Rivero MA, Encinoso M, Fernandez A. Osteonecrosis in pilot whales (Globicephala macrorhynchus). Res Vet Sci 2025; 192:105705. [PMID: 40412346 DOI: 10.1016/j.rvsc.2025.105705] [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: 03/07/2025] [Revised: 04/21/2025] [Accepted: 05/17/2025] [Indexed: 05/27/2025]
Abstract
Bone lesions, particularly osteonecrosis, have been rarely described in cetaceans, with limited studies correlating imaging, gross and microscopic findings. Aseptic osteonecrosis is especially relevant as a chronic form of decompression sickness, well-documented in human divers but controversially reported in cetaceans. This study examined the humeral heads of five stranded short-finned pilot whales (Globicephala macrorhynchus) using computed tomography (CT) and plastic embedding histologic techniques. Three specimens exhibited CT-detectable lesions, while two did not. CT imaging revealed distinct bone lesion patterns in three animals, including one case with a hypoattenuated triangular lesion constitutes histologically by well reorganized bone tissue. This lesion is highly consistent with aseptic dysbaric osteonecrosis (DON). Circular, focal, hyperattenuated necrotic bone areas most likely related to septic lesions were detected in two affected pilot whales. The two unaffected humoral heads exhibited normal bone histology, reinforcing the pathological significance of the osteonecrotic lesions of the three affected animals. As this was a retrospective study on fixed samples, microbiological analysis was not possible. Although not conclusive, this study provides the first histological evidence of chronic humeral head lesions in cetaceans consistent with DON, underscoring the need for further research using CT imaging, histology, and microbiology.
Collapse
Affiliation(s)
- Francesco Marie Achille Consoli
- Veterinary Histology and Pathology, Atlantic Centre for Cetacean Research (CAIC). University Institute of Animal Health and Food Safety (IUSA), WOAH col. Centre. Veterinary School, University of Las Palmas de Gran Canaria (ULPGC), Trasmontaña s/n, 35413 Arucas, Canary Islands, Spain
| | - Alonso Reyes-Matute
- Veterinary Histology and Pathology, Atlantic Centre for Cetacean Research (CAIC). University Institute of Animal Health and Food Safety (IUSA), WOAH col. Centre. Veterinary School, University of Las Palmas de Gran Canaria (ULPGC), Trasmontaña s/n, 35413 Arucas, Canary Islands, Spain; Faculty of Veterinary Medicine and Animal Husbandry (FMVZ), Pathology Department, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Miguel A Rivero
- Veterinary Histology and Pathology, Atlantic Centre for Cetacean Research (CAIC). University Institute of Animal Health and Food Safety (IUSA), WOAH col. Centre. Veterinary School, University of Las Palmas de Gran Canaria (ULPGC), Trasmontaña s/n, 35413 Arucas, Canary Islands, Spain
| | - Mario Encinoso
- Head of Imaging Diagnostic, Veterinary Hospital, Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, Trasmontaña, Arucas, 35413 Las Palmas, Spain
| | - Antonio Fernandez
- Veterinary Histology and Pathology, Atlantic Centre for Cetacean Research (CAIC). University Institute of Animal Health and Food Safety (IUSA), WOAH col. Centre. Veterinary School, University of Las Palmas de Gran Canaria (ULPGC), Trasmontaña s/n, 35413 Arucas, Canary Islands, Spain.
| |
Collapse
|
3
|
Yin Z, Huang D, Kuhn EMA, Moriarty TF, Li G, Wang X. Unraveling persistent bacteria: Formation, niches, and eradication strategies. Microbiol Res 2025; 297:128189. [PMID: 40311456 DOI: 10.1016/j.micres.2025.128189] [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: 03/12/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 05/03/2025]
Abstract
Persistent bacteria (persisters) are phenotypic variants that emerge either randomly or in response to a range of adverse environmental conditions. Persistence represents a state whereby a subpopulation of microorganisms can spontaneously enter a "dormant" state in response to environmental factors, while simultaneously exhibiting elevated tolerance to antimicrobial agents. This review provides the current definition of bacterial persistence and summarizes the mechanisms of persisters formation as well as the various niches of bacterial persistence encountered in clinical practice. Strategies targeting persisters are outlined, including but not limited to direct killing, awakening of persistent bacteria, combined clearance, and inhibition of persistence formation, and we conclude by proposing challenges and solutions for addressing bacterial persistence in current clinical practice.
Collapse
Affiliation(s)
- Zibo Yin
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China
| | - Diandian Huang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China
| | | | | | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China.
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029 PR China.
| |
Collapse
|
4
|
Mills EL, Suptela SR, Key MK, Marriott I, Johnson MB. RIG-I and cGAS mediate antimicrobial and inflammatory responses of primary osteoblasts and osteoclasts to Staphylococcus aureus. mBio 2025; 16:e0397124. [PMID: 40135931 PMCID: PMC12077190 DOI: 10.1128/mbio.03971-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Staphylococcus aureus is the primary causative agent of osteomyelitis, and it is now apparent that osteoblasts and osteoclasts play a significant role in the pathogenesis of such infections. Their responses can either be protective or exacerbate inflammatory bone loss and are mediated by the recognition of microbial motifs by various pattern recognition receptors. We have recently reported that osteoblasts can respond to S. aureus challenge with the production of the type I interferon, interferon-beta, which can reduce the number of viable bacteria harbored within infected cells. In the present study, we demonstrate that S. aureus viability and internalization are necessary for maximal inflammatory cytokine and type I interferon responses of primary bone cells to this pathogen. Importantly, we show that primary murine and human bone cells constitutively express the cytosolic nucleic acid sensors, retinoic acid inducible gene I (RIG-I) and cyclic GMP-AMP synthase (cGAS), and demonstrate that such expression is markedly upregulated following S. aureus infection. The functional status of RIG-I and cGAS in osteoblasts and osteoclasts was confirmed by showing that specific ligands for each can also elevate their expression and induce cytokine responses. We have verified the specificity of such responses using siRNA knockdown or pharmacological inhibition and used these approaches to demonstrate that both sensors play a pivotal role in bone cell responses to infection with clinically relevant strains of S. aureus. Finally, we have begun to establish the biological significance of RIG-I- and cGAS-mediated bone cell responses with the demonstration that their attenuation increases S. aureus burden in infected cells, suggesting a potentially protective role for these sensors in osteomyelitis.IMPORTANCEStaphylococcal osteomyelitis is a severe infection that is often recalcitrant to current treatment strategies. We and others have demonstrated that resident bone cells are not merely passive victims but can respond to bacteria with the production of an array of immune mediators, including type I interferons, that could serve to limit such infections. Here, we demonstrate the functional expression of two cytosolic nucleic acid sensors, retinoic acid inducible gene I and cyclic GMP-AMP synthase, in primary murine and human osteoblasts and murine osteoclasts. We show that these pattern recognition receptors mediate potentially protective bone cell type I interferon responses to Staphylococcus aureus infection.
Collapse
Affiliation(s)
- Erin L. Mills
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| | - Samantha R. Suptela
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| | - Mary-Kate Key
- Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, Georgia, USA
| | - Ian Marriott
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| | - M. Brittany Johnson
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina, USA
| |
Collapse
|
5
|
Song M, Sun J, Lv K, Li J, Shi J, Xu Y. A comprehensive review of pathology and treatment of staphylococcus aureus osteomyelitis. Clin Exp Med 2025; 25:131. [PMID: 40299136 PMCID: PMC12040984 DOI: 10.1007/s10238-025-01595-1] [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: 12/04/2024] [Accepted: 02/10/2025] [Indexed: 04/30/2025]
Abstract
Osteomyelitis (OM) is an inflammation of the bone and bone marrow triggered by infectious pathogens which may induce progressive bone destruction. The majority of OM cases, especially the chronic OM cases, are induced by the most prevalent and devastating pathogen Staphylococcus aureus (S. aureus), partially due to its resistance mechanisms against the immune system and antibiotic therapies. Regarding the high rate of morbidity and recurrence in patients, it is pivotal to elucidate underlying mechanisms that how S. aureus enter and survive in hosts. The accumulated discoveries have identified multiple distinct strategies associated with chronicity and recurrence include biofilm development, small colony variants (SCVs), staphylococcus abscess communities (SACs), the osteocyte lacuno-canalicular network invasion (OLCN) of cortical bones, and S. aureus protein A (SpA). Unfortunately, little clinical progress has been achieved for the diagnosis and therapeutic treatment for OM patients, indicating that numerous questions remain to be solved. Therefore, we still have a long way to obtain the clear elucidation of the host-pathogen interactions which could be applied for clinical treatment of OM. In this review, we provide insights of current knowledge about how S. aureus evades immune eradication and remains persistent in hosts with recent discoveries. The common and novel treatment strategies for OM are also described. The purpose of this review is to have in-dept understanding of S. aureus OM and bring new perspectives to therapeutic fields which may be translated to the clinic.
Collapse
Affiliation(s)
- Muguo Song
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Jian Sun
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Kehan Lv
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Junyi Li
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Jian Shi
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China.
| | - Yongqing Xu
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China.
| |
Collapse
|
6
|
Gao L, Tang Z, Zhang Z, Wei D, Wang J. Transcription factor FOS promotes ferroptosis and inflammation in S. aureus- infected osteomyelitis via EIF5A. J Orthop Surg Res 2025; 20:412. [PMID: 40270004 PMCID: PMC12020303 DOI: 10.1186/s13018-025-05815-y] [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: 10/16/2024] [Accepted: 04/12/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Osteomyelitis (OM) is a bone disease that can leave people disabled. Eukaryotic translation initiation factor (EIF5A) is involved in cell proliferation, apoptosis, differentiation, and inflammation, but the role of EIF5A in staphylococcus aureus (S. aureus)-infected OM remains unclear. METHODS The mRNA and proteins were detected by qRT-PCR and western blot. Cell viability was examined by CCK8 assay. The reactive oxygen species (ROS), malondialdehyde (MDA), ferrous iron (Fe2+), and glutathione (GSH) levels were analyzed using the ROS, MDA, GSH, and Fe2+ detection kits. The levels of tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and Interleukin-6 (IL-6) were examined using Enzyme-linked immunosorbent (ELISA) kits. The binding between FOS and promoter of EIF5A was detected by chromatin immunoprecipitation (CHIP) assay. The interaction between EIF5A and Fos proto-oncogene (FOS) was detected by dual-luciferase reporter assay. The diagnostic values of EIF5A and FOS were analyzed with blood of S. aureus-infected OM patients and healthy volunteers by ROC curve. RESULTS The EIF5A was up-regulated in S. aureus-infected OM. EIF5A knockdown promoted cell viability in S. aureus-infected MG-63 cells and reduced ROS, MDA, and Fe2+ levels, and increased GSH levels. Meanwhile, silencing EIF5A could increase expression of glutathione peroxidase 4 (GPX4), and ferritin heavy chain1 (FTH1) and reduce acyl-CoA synthetase long-chain family member 4 (ACSL4) expression, and silencing EIF5A could reduce immune factors (TNF-α, IL-1β, and IL-6) levels. FOS could bind to EIF5A. Silencing FOS promoted cell viability, and increased GSH levels in S. aureus-infected MG-63 cells, but reduced ROS, MDA, and Fe2+ levels. Meanwhile, promoted GPX4 and FTH1 expression, inhibited ACSL4 expression, and reduced immune factor levels in S. aureus-infected MG-63 cells. Interestingly, EIF5A overexpression could weaken the actions. FOS promotes ferroptosis and inflammation via EIF5A in S. aureus-infected MG-63 cells. Besides, the EIF5A and FOS might be potential molecular diagnostic markers in the progression of OM. CONCLUSION FOS promotes ferroptosis and inflammation via EIF5A in S. aureus-infected OM. This study is first to report the role of FOS and EIF5A in S. aureus-infected OM, but we found that there are still some limitations in our work, such as not covering all possible types of infection, which is the focus of future research.
Collapse
Affiliation(s)
- Lei Gao
- Orthopedic Department, Capital Medical University Affiliated Beijing Shijitan Hospital, No.10 Yangfangdian Tieyi Road, Haidian District, Beijing, China
| | - Zhipeng Tang
- Clinnical Laboratory, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Zhijin Zhang
- Department of Hand Surgery, Jiangnan Hospital Affiliated Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Dehua Wei
- Orthopedic Department, Capital Medical University Affiliated Beijing Shijitan Hospital, No.10 Yangfangdian Tieyi Road, Haidian District, Beijing, China
| | - Jiangning Wang
- Orthopedic Department, Capital Medical University Affiliated Beijing Shijitan Hospital, No.10 Yangfangdian Tieyi Road, Haidian District, Beijing, China.
| |
Collapse
|
7
|
Elgharably H, Claesen J, Sangwan N, Etiwy M, Houghtaling P, Procop GW, Shrestha NK, Griffin B, Navia JL, Svensson LG, Wozniak DJ, Pettersson GB. In vivo virulence of Staphylococcus aureus in native versus prosthetic left-sided valve endocarditis. JTCVS OPEN 2025; 24:156-169. [PMID: 40309682 PMCID: PMC12039428 DOI: 10.1016/j.xjon.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/23/2024] [Accepted: 12/03/2024] [Indexed: 05/02/2025]
Abstract
Objectives Staphylococcus aureus infective endocarditis is commonly associated with invasive pathology and is worse in prosthetic valve endocarditis. In this study, we aim to examine S. aureus virulence and pathological features of native and prosthetic valve infective endocarditis. Methods Between 2002 and 2020, 438 patients underwent surgery for left-sided endocarditis caused by S. aureus at our center (59% native and 41% prosthetic valve endocarditis). Endocarditis registry was queried, and pathological features were based on the echocardiography and operative findings. In addition, vegetation samples were collected from 6 patients undergoing surgery for infective endocarditis (3 native and 3 prosthetic valve endocarditis). Total RNA was extracted from all specimens, and messenger RNA sequencing was executed for transcriptomic analysis. Data were pooled into STAR aligner, and gene expression related to virulence factors was compared between 2 groups. Results Rates of invasive pathology were higher in prosthetic versus native valve infective endocarditis (76% vs 40%, P < .0001), which impacted the complexity of surgical procedures and perioperative course, but not in-hospital mortality. Transcriptomic analysis has shown differences in gene expression between vegetation specimens of native and prosthetic valve endocarditis, including genes for stress response, biofilm formation, and virulence factors. The gene aur (encodes for aureolysin) was highly upregulated in prosthetic valve vegetations compared with native valve vegetations (P = .023). Conclusions Prosthetic valve endocarditis caused by S. aureus is associated with further invasive pathology compared with native valve endocarditis, which could be related to upregulation of genes responsible for biofilm formation and metalloproteinase production.
Collapse
Affiliation(s)
- Haytham Elgharably
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, Ohio
| | - Jan Claesen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Naseer Sangwan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Microbial Sequencing & Analytics Resource (MSAAR) Facility, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Muhammad Etiwy
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, Ohio
| | - Penny Houghtaling
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, Ohio
| | - Gary W. Procop
- Department Laboratory Medicine, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Brian Griffin
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Jose L. Navia
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, Ohio
| | - Lars G. Svensson
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, Ohio
| | - Daniel J. Wozniak
- Departments of Microbial Infection and Immunity and Microbiology, Ohio State University, Columbus, Ohio
| | - Gosta B. Pettersson
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
8
|
Denina M, Abrate G, Silvestro E, Funiciello E, Pruccoli G, Sandei M, Mazzetti G, Garazzino S. Daptomycin for Pediatric Complex Bone and Joint Infections: Real-world Efficacy and Safety Data from a Three-year Study. Pediatr Infect Dis J 2025:00006454-990000000-01262. [PMID: 40106774 DOI: 10.1097/inf.0000000000004804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Bone and joint infections present a significant therapeutic challenge in children, particularly in complex or chronic cases. Staphylococcus aureus is the most common causative pathogen, with methicillin resistance rates varying by geographic location and hospital setting, underscoring the need for antibiotics effective against both methicillin-sensitive Staphylococcus aureus and methicillin-resistant Staphylococcus aureus. Daptomycin, a cyclic lipopeptide antibiotic effective against gram-positive pathogens, is increasingly used off-label in pediatric osteomyelitis. This prospective study evaluates its real-world application, focusing on clinical outcomes, dosing strategies, safety, and therapeutic potential. Over a 3-year period, 20 children (median age: 13.5 years) with bone and joint infections were treated with daptomycin. The patient cohort was highly complex, with 70% having severe orthopedic or medical comorbidities. Methicillin-sensitive Staphylococcus aureus was isolated in 60% of cases, while MRSA was identified in 35%. Daptomycin was administered following bacteremia dosing guidelines, with some patients receiving higher doses (mean: 10 mg/kg). The median duration of daptomycin therapy was 24.5 days. Treatment was successful in 90% of cases, often transitioning to oral antibiotic therapy. C-reactive protein levels showed a median reduction of 90%. A composite score evaluating clinical response-including C-reactive protein levels, type of continuation therapy, and the need for surgery-indicated substantial improvement in the majority of patients, with a median score of 4. No severe adverse events were recorded, though transient neutropenia and elevated creatine kinase levels were observed in 2 cases. This study underscores daptomycin's safety and efficacy in treating complex pediatric osteoarticular infections and suggests its potential as a first-line therapy, particularly for MRSA cases. The comprehensive data reflect the current microbiological landscape of pediatric bone and joint infections, supporting the reconsideration of daptomycin's role in initial treatment protocols. Further research and controlled trials are warranted to confirm these findings and optimize treatment strategies.
Collapse
Affiliation(s)
- Marco Denina
- From the Department of Pediatrics, Infectious Diseases Unit, University of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Giulia Abrate
- Department of Pediatrics and Public Health, University of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Erika Silvestro
- From the Department of Pediatrics, Infectious Diseases Unit, University of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Elisa Funiciello
- Department of Pediatric Onco-Hematology, University of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Giulia Pruccoli
- From the Department of Pediatrics, Infectious Diseases Unit, University of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Matteo Sandei
- Department of Pediatrics and Public Health, University of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Giulia Mazzetti
- Department of Pediatrics and Public Health, University of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Silvia Garazzino
- From the Department of Pediatrics, Infectious Diseases Unit, University of Turin, Regina Margherita Children's Hospital, Turin, Italy
| |
Collapse
|
9
|
Varughese J, Halfman A, Crotty M, Alexander J, Hunter L, Hupert M, Dominguez E. Do not treat ghosts: anti-methicillin-resistant Staphylococcus aureus (MRSA) therapy in osteomyelitis without identified MRSA. ANTIMICROBIAL STEWARDSHIP & HEALTHCARE EPIDEMIOLOGY : ASHE 2025; 5:e53. [PMID: 40026767 PMCID: PMC11869046 DOI: 10.1017/ash.2025.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/31/2024] [Accepted: 01/01/2025] [Indexed: 03/05/2025]
Abstract
Objective To compare the clinical outcomes of patients with lower limb osteomyelitis (LLOM) and negative methicillin-resistant Staphylococcus aureus (MRSA) cultures treated with anti-MRSA therapy (AMT) versus those treated with no-anti-MRSA therapy (NAMT). Design Retrospective cohort study. Patients Hospitalized adult (≥18 yr of age) patients admitted to multiple tertiary referral centers in a single healthcare system between April 1, 2017 and April 1, 2023, with LLOM and planned intravenous antibiotics for at least four weeks. Methods Electronic medical records were queried for demographic information, admission dates, treatment strategies, imaging and culture results, and discharge diagnoses. Descriptive statistics measured baseline characteristics, imaging, and culture results. Results Out of 473 patients, 64 met the inclusion criteria and 409 were excluded. Of the 64 patients, 26 (40%) had AMT and 38 (59%) had NAMT. A larger but statistically insignificant portion of patients in the NAMT cohort failed therapy (23% AMT vs 32% NAMT, P = 0.325). However, hospital readmission for LLOM within 180 days of antibiotic completion (46.2% vs 47%, P = 0.92), hospital length of stay (median (IQR): 6 (5-9) d vs 7 (5-12.5) d, P = 0.285), incidence of new renal replacement therapy initiation (0% vs 2.6%, P = 0.594), creatinine kinase levels (0 vs 2.6%, P = 0.594), and drug-induced immune thrombocytopenia (0% vs 5.3% P = 0.349) were comparable between the two cohorts. Conclusions Treatment failure rates and adverse events did not differ significantly among patients with LLOM treated with AMT or NAMT. Further investigation of determinants of clinical failures in LLOM may help optimize overall treatment.
Collapse
Affiliation(s)
- Jincy Varughese
- Department of Pharmacy, Methodist Dallas Medical Center, Dallas, TX, USA
| | - Annie Halfman
- Department of Pharmacy, Methodist Dallas Medical Center, Dallas, TX, USA
| | - Matthew Crotty
- Department of Pharmacy, Methodist Dallas Medical Center, Dallas, TX, USA
| | - Julie Alexander
- Department of Infectious Diseases, Methodist Dallas Medical Center, Dallas, TX, USA
| | - Leigh Hunter
- Department of Internal Medicine, Methodist Dallas Medical Center, Dallas, TX, USA
| | - Mark Hupert
- Department of Infectious Diseases, Methodist Dallas Medical Center, Dallas, TX, USA
| | - Edward Dominguez
- Department of Infectious Diseases, Methodist Dallas Medical Center, Dallas, TX, USA
- The Liver Institute at Methodist Dallas, Methodist Dallas Medical Center, Dallas, TX, USA
| |
Collapse
|
10
|
Dong Q, Zhou J, Feng M, Kong L, Fang B, Zhang Z. A review of bacterial and osteoclast differentiation in bone infection. Microb Pathog 2024; 197:107102. [PMID: 39505086 DOI: 10.1016/j.micpath.2024.107102] [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: 03/29/2024] [Revised: 10/18/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
Bone infections are characterized by bacterial invasion of the bone microenvironment and subsequent bone structure deterioration. This holds significance because osteoclasts, which are the only cells responsible for bone resorption, are abnormally stimulated during bone infections. Multiple communication factors secreted by bone stromal cells regulate the membrane of osteoclast progenitor cells, thereby maintaining bone homeostasis through the expression of many types of receptors. During infection, the immunoinflammatory response triggered by bacterial invasion and multiple virulence factors of bacterial origin can disrupt osteoclast homeostasis. Therefore, clarifying the pathways through which bacteria affect osteoclasts can offer a theoretical basis for preventing and treating bone infections. This review summarizes studies investigating bone destruction caused by different bacterial infections. In conclusion, bacteria can affect osteoclast metabolic activity through multiple pathways, including direct contact, release of virulence factors, induction of immunoinflammatory responses, influence on bone stromal cell metabolism, and intracellular infections.
Collapse
Affiliation(s)
- Qi Dong
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Jiuqin Zhou
- Department of Infectious Disease of Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Mingzhe Feng
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Lingqiang Kong
- Department of Orthopedics, the Central Hospital Affiliated to Shaoxing University, Shaoxing, 312030, China.
| | - Bin Fang
- Department of Orthopedics, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310000, China.
| | - Zhen Zhang
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China.
| |
Collapse
|
11
|
Mayer P, Smith AC, Hurlow J, Morrow BR, Bohn GA, Bowler PG. Assessing Biofilm at the Bedside: Exploring Reliable Accessible Biofilm Detection Methods. Diagnostics (Basel) 2024; 14:2116. [PMID: 39410520 PMCID: PMC11475494 DOI: 10.3390/diagnostics14192116] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 10/20/2024] Open
Abstract
INTRODUCTION Biofilm is linked through a variety of mechanisms to the pathogenesis of chronic wounds. However, accurate biofilm detection is challenging, demanding highly specialized and technically complex methods rendering it unapplicable for most clinical settings. This study evaluated promising methods of bedside biofilm localization, fluorescence imaging of wound bacterial loads, and biofilm blotting by comparing their performance against validation scanning electron microscopy (SEM). METHODS In this clinical trial, 40 chronic hard-to-heal wounds underwent the following assessments: (1) clinical signs of biofilm (CSB), (2) biofilm blotting, (3) fluorescence imaging for localizing bacterial loads, wound scraping taken for (4) SEM to confirm matrix encased bacteria (biofilm), and (5) PCR (Polymerase Chain Reaction) and NGS (Next Generation Sequencing) to determine absolute bacterial load and species present. We used a combination of SEM and PCR microbiology to calculate the diagnostic accuracy measures of the CSB, biofilm blotting assay, and fluorescence imaging. RESULTS Study data demonstrate that 62.5% of wounds were identified as biofilm-positive based on SEM and microbiological assessment. By employing this method to determine the gold truth, and thus calculate accuracy measures for all methods, fluorescence imaging demonstrated superior sensitivity (84%) and accuracy (63%) compared to CSB (sensitivity 44% and accuracy 43%) and biofilm blotting (sensitivity 24% and accuracy 40%). Biofilm blotting exhibited the highest specificity (64%), albeit with lower sensitivity and accuracy. Using SEM alone as the validation method slightly altered the results, but all trends held constant. DISCUSSION This trial provides the first comparative assessment of bedside methods for wound biofilm detection. We report the diagnostic accuracy measures of these more feasibly implementable methods versus laboratory-based SEM. Fluorescence imaging showed the greatest number of true positives (highest sensitivity), which is clinically relevant and provides assurance that no pathogenic bacteria will be missed. It effectively alerted regions of biofilm at the point-of-care with greater accuracy than standard clinical assessment (CSB) or biofilm blotting paper, providing actionable information that will likely translate into enhanced therapeutic approaches and better patient outcomes.
Collapse
Affiliation(s)
- Perry Mayer
- The Mayer Institute (TMI), Hamilton, ON L8R 2R3, Canada
| | - Allie Clinton Smith
- Department of Honors Studies, Texas Tech University, Lubbock, TX 79409, USA;
| | - Jennifer Hurlow
- Consultant Wound Care Specialized Nurse Practitioner, Memphis, TN 38120, USA;
| | - Brian R. Morrow
- College of Dentistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Gregory A. Bohn
- The American Professional Wound Care Association (APWCA), American Board of Wound Healing, Milwaukee, WI 53214, USA
| | | |
Collapse
|
12
|
Zhang Y, Chen J, Sun Y, Wang M, Liu H, Zhang W. Endogenous Tissue Engineering for Chondral and Osteochondral Regeneration: Strategies and Mechanisms. ACS Biomater Sci Eng 2024; 10:4716-4739. [PMID: 39091217 DOI: 10.1021/acsbiomaterials.4c00603] [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: 08/04/2024]
Abstract
Increasing attention has been paid to the development of effective strategies for articular cartilage (AC) and osteochondral (OC) regeneration due to their limited self-reparative capacities and the shortage of timely and appropriate clinical treatments. Traditional cell-dependent tissue engineering faces various challenges such as restricted cell sources, phenotypic alterations, and immune rejection. In contrast, endogenous tissue engineering represents a promising alternative, leveraging acellular biomaterials to guide endogenous cells to the injury site and stimulate their intrinsic regenerative potential. This review provides a comprehensive overview of recent advancements in endogenous tissue engineering strategies for AC and OC regeneration, with a focus on the tissue engineering triad comprising endogenous stem/progenitor cells (ESPCs), scaffolds, and biomolecules. Multiple types of ESPCs present within the AC and OC microenvironment, including bone marrow-derived mesenchymal stem cells (BMSCs), adipose-derived mesenchymal stem cells (AD-MSCs), synovial membrane-derived mesenchymal stem cells (SM-MSCs), and AC-derived stem/progenitor cells (CSPCs), exhibit the ability to migrate toward injury sites and demonstrate pro-regenerative properties. The fabrication and characteristics of scaffolds in various formats including hydrogels, porous sponges, electrospun fibers, particles, films, multilayer scaffolds, bioceramics, and bioglass, highlighting their suitability for AC and OC repair, are systemically summarized. Furthermore, the review emphasizes the pivotal role of biomolecules in facilitating ESPCs migration, adhesion, chondrogenesis, osteogenesis, as well as regulating inflammation, aging, and hypertrophy-critical processes for endogenous AC and OC regeneration. Insights into the applications of endogenous tissue engineering strategies for in vivo AC and OC regeneration are provided along with a discussion on future perspectives to enhance regenerative outcomes.
Collapse
Affiliation(s)
- Yanan Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Mingyue Wang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| |
Collapse
|
13
|
Shi S, Shi W, Zhou B, Qiu S. Research and Application of Chitosan Nanoparticles in Orthopedic Infections. Int J Nanomedicine 2024; 19:6589-6602. [PMID: 38979535 PMCID: PMC11228078 DOI: 10.2147/ijn.s468848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/15/2024] [Indexed: 07/10/2024] Open
Abstract
Orthopedic infection is one of the most intractable orthopedic problems. Bacteria resistant to antibiotics also develop gradually. Chitosan is widely used in the Biomedical field because of its high biocompatibility, biodegradability, and antibacterial activity. Chitosan-based drug delivery systems are frequently utilized to produce controlled medication release. When combined with antibiotics, synergistic antibacterial effects can be achieved. Chitosan-based nanoparticles are one of the most widely used applications in drug delivery systems. The focus of this review is to provide information on new methods being developed for chitosan-based nanoparticles in the field of bone infection treatment, including chitosan nanoparticles for antibacterial purposes, Ch-loaded with antibiotics, Ch-loaded with metal, and used as immune adjuvants. It may Provide ideas for the fundamental research and the prospects of future clinical applications of orthopedic infections.
Collapse
Affiliation(s)
- Sifeng Shi
- Department of Orthopedic Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Weiran Shi
- Department of Orthopedic Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Bing Zhou
- Department of Orthopedic Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Shang Qiu
- Department of Orthopedic Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| |
Collapse
|
14
|
Xia Z, Liao Y, Gao G, Zhang S. Rifampicin-Loaded Polyelectrolyte Complex Eliminates Intracellular Bacteria through Thiol-Mediated Cellular Uptake and Oxidative Stress Enhancement. ACS APPLIED BIO MATERIALS 2024; 7:2544-2553. [PMID: 38507285 DOI: 10.1021/acsabm.4c00143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
The poor accumulation of antibiotics in the cytoplasm leads to the poor eradication of intracellular bacteria. Herein, a polyelectrolyte complex (PECs@Rif) allowing direct cytosolic delivery of rifampicin (Rif) was developed for the treatment of intracellular infections by complexation of poly(α-lipoic acid) (pLA) and oligosaccharide (COS) in water and loading Rif. Due to the thiol-mediated cellular uptake, PECs@Rif delivered 3.9 times higher Rif into the cytoplasm than that of the free Rif during 8 h of incubation. After entering cells, PECs@Rif released Rif by dissociating pLA into dihydrolipoic acid (DHLA) in the presence of intracellular thioredoxin reductase (TrxR). Notably, DHLA could reduce endogenous Fe(III) to Fe(II) and provide a catalyst for the Fenton reaction to produce a large amount of reactive oxygen species (ROS), which would assist Rif in eradicating intracellular bacteria. In vitro assay showed that PECs@Rif reduced almost 2.8 orders of magnitude of intracellular bacteria, much higher than 0.7 orders of magnitude of free Rif. The bacteremia-bearing mouse models showed that PECs@Rif reduced bacterial levels in the liver, spleen, and kidney by 2.2, 3.7, and 2.3 orders of magnitude, respectively, much higher than free Rif in corresponding tissues. The direct cytosolic delivery in a thiol-mediated manner and enhanced oxidative stress proposed a feasible strategy for treating intracellular bacteria infection.
Collapse
Affiliation(s)
- Zhaoxin Xia
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Yulong Liao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Ge Gao
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Shiyong Zhang
- College of Chemistry, Sichuan University, Chengdu 610064, China
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
15
|
Hajdu KS, Baker CE, Moore-Lotridge SN, Schoenecker JG. Sequestration and Involucrum: Understanding Bone Necrosis and Revascularization in Pediatric Orthopedics. Orthop Clin North Am 2024; 55:233-246. [PMID: 38403369 DOI: 10.1016/j.ocl.2023.09.005] [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: 02/27/2024]
Abstract
Sequestration, a condition where a section of bone becomes necrotic due to a loss of vascularity or thrombosis, can be a challenging complication of osteomyelitis. This review explores the pathophysiology of sequestration, highlighting the role of the periosteum in forming involucrum and creeping substitution which facilitate revascularization and bone formation. The authors also discuss the induced membrane technique, a two-stage surgical procedure for cases of failed healing of sequestration. Future directions include the potential use of prophylactic anticoagulation and novel drugs targeting immunocoagulopathy, as well as the development of advanced imaging techniques and single-stage surgical procedures.
Collapse
Affiliation(s)
- Katherine S Hajdu
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Courtney E Baker
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Stephanie N Moore-Lotridge
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jonathan G Schoenecker
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee, USA.
| |
Collapse
|
16
|
Chen Z, Wang L, He D, Liu Q, Han Q, Zhang J, Zhang AM, Song Y. Exploration of the Antibacterial and Anti-Inflammatory Activity of a Novel Antimicrobial Peptide Brevinin-1BW. Molecules 2024; 29:1534. [PMID: 38611812 PMCID: PMC11013252 DOI: 10.3390/molecules29071534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Antibiotic resistance has emerged as a grave threat to global public health, leading to an increasing number of treatment failures. Antimicrobial peptides (AMPs) are widely regarded as potential substitutes for traditional antibiotics since they are less likely to induce resistance when used. A novel AMP named Brevinin-1BW (FLPLLAGLAASFLPTIFCKISRKC) was obtained by the Research Center of Molecular Medicine of Yunnan Province from the skin of the Pelophylax nigromaculatus. Brevinia-1BW had effective inhibitory effects on Gram-positive bacteria, with a minimum inhibitory concentration (MIC) of 3.125 μg/mL against Enterococcus faecalis (ATCC 29212) and 6.25 μg/mL against both Staphylococcus aureus (ATCC 25923) and multidrug-resistant Staphylococcus aureus (ATCC 29213) but had weaker inhibitory effects on Gram-negative bacteria, with a MIC of ≥100 μg/mL. Studies using scanning electron microscopy (SEM) and flow cytometry have revealed that it exerts its antibacterial activity by disrupting bacterial membranes. Additionally, it possesses strong biofilm inhibitory and eradication activities as well as significant lipopolysaccharide (LPS)-binding activity. Furthermore, Brevinin-1BW has shown a significant anti-inflammatory effect in LPS-treated RAW264.7 cells. In conclusion, Brevinin-1BW is anticipated to be a promising clinical agent with potent anti-Gram-positive bacterial and anti-inflammatory properties.
Collapse
Affiliation(s)
- Zhizhi Chen
- Research Center of Molecular Medicine of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650504, China; (Z.C.); (L.W.); (D.H.); (Q.L.); (Q.H.); (J.Z.); (A.-M.Z.)
| | - Lei Wang
- Research Center of Molecular Medicine of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650504, China; (Z.C.); (L.W.); (D.H.); (Q.L.); (Q.H.); (J.Z.); (A.-M.Z.)
| | - Dongxia He
- Research Center of Molecular Medicine of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650504, China; (Z.C.); (L.W.); (D.H.); (Q.L.); (Q.H.); (J.Z.); (A.-M.Z.)
| | - Qi Liu
- Research Center of Molecular Medicine of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650504, China; (Z.C.); (L.W.); (D.H.); (Q.L.); (Q.H.); (J.Z.); (A.-M.Z.)
| | - Qinqin Han
- Research Center of Molecular Medicine of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650504, China; (Z.C.); (L.W.); (D.H.); (Q.L.); (Q.H.); (J.Z.); (A.-M.Z.)
| | - Jinyang Zhang
- Research Center of Molecular Medicine of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650504, China; (Z.C.); (L.W.); (D.H.); (Q.L.); (Q.H.); (J.Z.); (A.-M.Z.)
| | - A-Mei Zhang
- Research Center of Molecular Medicine of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650504, China; (Z.C.); (L.W.); (D.H.); (Q.L.); (Q.H.); (J.Z.); (A.-M.Z.)
| | - Yuzhu Song
- Research Center of Molecular Medicine of Yunnan Province, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650504, China; (Z.C.); (L.W.); (D.H.); (Q.L.); (Q.H.); (J.Z.); (A.-M.Z.)
- School of Medicine, Kunming University of Science and Technology, Kunming 650504, China
| |
Collapse
|
17
|
Hussain MS, Shaikh NK, Agrawal M, Tufail M, Bisht AS, Khurana N, Kumar R. Osteomyelitis and non-coding RNAS: A new dimension in disease understanding. Pathol Res Pract 2024; 255:155186. [PMID: 38350169 DOI: 10.1016/j.prp.2024.155186] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/27/2024] [Accepted: 01/31/2024] [Indexed: 02/15/2024]
Abstract
Osteomyelitis, a debilitating bone infection, presents considerable clinical challenges due to its intricate etiology and limited treatment options. Despite strides in surgical and chemotherapeutic interventions, the treatment landscape for osteomyelitis remains unsatisfactory. Recent attention has focused on the role of non-coding RNAs (ncRNAs) in the pathogenesis and progression of osteomyelitis. This review consolidates current knowledge on the involvement of distinct classes of ncRNAs, including microRNAs, long ncRNAs, and circular RNAs, in the context of osteomyelitis. Emerging evidence from various studies underscores the potential of ncRNAs in orchestrating gene expression and influencing the differentiation of osteoblasts and osteoclasts, pivotal processes in bone formation. The review initiates by elucidating the regulatory functions of ncRNAs in fundamental cellular processes such as inflammation, immune response, and bone remodeling, pivotal in osteomyelitis pathology. It delves into the intricate network of interactions between ncRNAs and their target genes, illuminating how dysregulation contributes to the establishment and persistence of osteomyelitic infections. Understanding their regulatory roles may pave the way for targeted diagnostic tools and innovative therapeutic interventions, promising a paradigm shift in the clinical approach to this challenging condition. Additionally, we delve into the promising therapeutic applications of these molecules, envisioning novel diagnostic and treatment approaches to enhance the management of this challenging bone infection.
Collapse
Affiliation(s)
- Md Sadique Hussain
- Department of Pharmacology, School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan 302017, India
| | - Nusrat K Shaikh
- Department of Quality Assurance, Smt. N. M. Padalia Pharmacy College, Ahmedabad, 382210 Gujarat, India
| | - Mohit Agrawal
- Department of Pharmacology, School of Medical & Allied Sciences, K.R. Mangalam University, Gurugram 122103, India
| | - Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.
| | - Ajay Singh Bisht
- School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Patel Nagar, Dehradun, Uttarakhand 248001, India
| | - Navneet Khurana
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rajesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
18
|
Baharvand R, Fallah F, Jafari P, Azimi L. Co-colonization of methicillin-resistant Staphylococcus aureus and Candida spp. in children with malignancies. AMB Express 2024; 14:22. [PMID: 38351284 PMCID: PMC10864235 DOI: 10.1186/s13568-024-01667-7] [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: 06/18/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
This study aimed to evaluate the interaction between methicillin-resistant Staphylococcus aureus(MRSA) and Candida spp. in the oral cavity of children with malignancies under chemotherapy. We evaluated the expression level of Als3p and mecA in Candida spp. and MRSA strains in both single colonization and co-colonization condition. Oral and nasal samples were collected by dry sponge swabs in 10 ml of sterile phosphate-buffered saline. The MRSA and Candida spp. was confirmed using the PCR method and mecA and Als3p genes, respectively. The SYBR Green-based quantitative real-time PCR was used to evaluate the relative expression levels of mecA and Als3p genes in MRSA and Candida spp., respectively. The frequency of S. aureus in oral-only and nasal-only swab samples were 14.1% (n = 24/170). 58.3% (n = 14/24) and 29.2% (n = 7/24) of S. aureus isolated from oral and nasal samples were MRSA, respectively. Among Candida species, C. albicans (n = 28/170; 16.5%) had the highest frequency. The oral co-colonization of MRSA and Candida spp. was detected in 4.7% (n = 8/170) patients. The overall average of gene expression levels among all Candida spp. and MRSA isolates indicated that the mecA and Als3p genes expression increased six and two times in co-colonization conditions compared to single colonization conditions, respectively. Our findings revealed the importance of polymicrobial infection in clinical settings and stated that it is possible that Candida spp. facilitates the infection of S. aureus and can lead to systemic infection in co-colonized patients.
Collapse
Affiliation(s)
- Raziyeh Baharvand
- Department of Microbiology, Faculty of Science, Islamic Azad University, Arak branch, Arak, Iran
| | - Fatemeh Fallah
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvaneh Jafari
- Department of Microbiology, Faculty of Science, Islamic Azad University, Arak branch, Arak, Iran.
| | - Leila Azimi
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Doub JB, Heil EL, Manson T. Adjuvant intra-articular vancomycin for recalcitrant Staphylococcal prosthetic joint infections of the knee. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY & TRAUMATOLOGY : ORTHOPEDIE TRAUMATOLOGIE 2024; 34:1031-1036. [PMID: 37864658 DOI: 10.1007/s00590-023-03764-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/09/2023] [Indexed: 10/23/2023]
Abstract
OBJECTIVE Chronic prosthetic joint infection patients who fail conventional two-stage revision surgery are an especially difficult to treat patient population. Consequently, the objective of this study was to investigate the safety and long-term effectiveness of adjuvant intra-articular vancomycin therapy in conjunction with two-stage revision knee arthroplasties for recalcitrant Staphylococcal prosthetic joint infections. METHODS This was an observational cohort study of twelve patients with recalcitrant Staphylococcal prosthetic joint infections of the knee which had failed previous revision surgeries. Each patient subsequently underwent two-stage revision with placement of Hickman catheters to deliver intra-articular vancomycin therapy. In addition, systemic antibiotic therapy was administered for 6 weeks, and long-term follow-up was evaluated then for 5 years. RESULTS Seventy-five percent of the cohort have had no recurrence of their infections at 5 years. Two patients formed fistulas requiring above the knee amputations, and three patients had acute kidney injury. All patients had maximum measurable serum vancomycin trough levels that ranged from 6.1 to 93.6 mcg/mL. CONCLUSION The aggressive protocol used in this cohort with repeat two-stage revision surgery, intra-articular vancomycin and systemic antibiotics was able to prevent recurrence of infection in most patients, but higher than expected rates of acute kidney injury were observed in this study. Therefore, while intra-articular vancomycin therapy may have some effectiveness in treating recalcitrant prosthetic joint infections, its ability to eradicate all bacterial niduses is unproven, and clinicians should be cognizant of potential adverse events that can occur with this therapy.
Collapse
Affiliation(s)
- James B Doub
- Division of Infectious Diseases, University of Maryland School of Medicine, 725 West Lombard Street, BaltimoreMaryland, MD, 21201, USA.
| | - Emily L Heil
- Department of Pharmacy Practice and Science, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Theodore Manson
- Department of Orthopedic Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
20
|
Alfei S. Shifting from Ammonium to Phosphonium Salts: A Promising Strategy to Develop Next-Generation Weapons against Biofilms. Pharmaceutics 2024; 16:80. [PMID: 38258091 PMCID: PMC10819902 DOI: 10.3390/pharmaceutics16010080] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Since they are difficult and sometimes impossible to treat, infections sustained by multidrug-resistant (MDR) pathogens, emerging especially in nosocomial environments, are an increasing global public health concern, translating into high mortality and healthcare costs. In addition to having acquired intrinsic abilities to resist available antibiotic treatments, MDR bacteria can transmit genetic material encoding for resistance to non-mutated bacteria, thus strongly decreasing the number of available effective antibiotics. Moreover, several pathogens develop resistance by forming biofilms (BFs), a safe and antibiotic-resistant home for microorganisms. BFs are made of well-organized bacterial communities, encased and protected in a self-produced extracellular polymeric matrix, which impedes antibiotics' ability to reach bacteria, thus causing them to lose efficacy. By adhering to living or abiotic surfaces in healthcare settings, especially in intensive care units where immunocompromised older patients with several comorbidities are hospitalized BFs cause the onset of difficult-to-eradicate infections. In this context, recent studies have demonstrated that quaternary ammonium compounds (QACs), acting as membrane disruptors and initially with a low tendency to develop resistance, have demonstrated anti-BF potentialities. However, a paucity of innovation in this space has driven the emergence of QAC resistance. More recently, quaternary phosphonium salts (QPSs), including tri-phenyl alkyl phosphonium derivatives, achievable by easy one-step reactions and well known as intermediates of the Wittig reaction, have shown promising anti-BF effects in vitro. Here, after an overview of pathogen resistance, BFs, and QACs, we have reviewed the QPSs developed and assayed to this end, so far. Finally, the synthetic strategies used to prepare QPSs have also been provided and discussed to spur the synthesis of novel compounds of this class. We think that the extension of the knowledge about these materials by this review could be a successful approach to finding effective weapons for treating chronic infections and device-associated diseases sustained by BF-producing MDR bacteria.
Collapse
Affiliation(s)
- Silvana Alfei
- Department of Pharmacy, University of Genoa, Viale Cembrano, 4, 16148 Genova, Italy
| |
Collapse
|
21
|
Campbell MJ, Beenken KE, Ramirez AM, Smeltzer MS. The major role of sarA in limiting Staphylococcus aureus extracellular protease production in vitro is correlated with decreased virulence in diverse clinical isolates in osteomyelitis. Virulence 2023; 14:2175496. [PMID: 36748843 PMCID: PMC9928472 DOI: 10.1080/21505594.2023.2175496] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
We previously demonstrated that MgrA, SarA, SarR, SarS, SarZ, and Rot bind at least three of the four promoters associated with genes encoding primary extracellular proteases in Staphylococcus aureus (Aur, ScpA, SspA/SspB, SplA-F). We also showed that mutation of sarA results in a greater increase in protease production, and decrease in biofilm formation, than mutation of the loci encoding any of these other proteins. However, these conclusions were based on in vitro studies. Thus, the goal of the experiments reported here was to determine the relative impact of the regulatory loci encoding these proteins in vivo. To this end, we compared the virulence of mgrA, sarA, sarR, sarS, sarZ, and rot mutants in a murine osteomyelitis model. Mutants were generated in the methicillin-resistant USA300 strain LAC and the methicillin-sensitive USA200 strain UAMS-1, which was isolated directly from the bone of an osteomyelitis patient during surgical debridement. Mutation of mgrA and rot limited virulence to a statistically significant extent in UAMS-1, but not in LAC, while the sarA mutant exhibited reduced virulence in both strains. The reduced virulence of the sarA mutant was correlated with reduced cytotoxicity for osteoblasts and osteoclasts, reduced biofilm formation, and reduced sensitivity to the antimicrobial peptide indolicidin, all of which were directly attributable to increased protease production in both LAC and UAMS-1. These results illustrate the importance of considering diverse clinical isolates when evaluating the impact of regulatory mutations on virulence and demonstrate the significance of SarA in limiting protease production in vivo in S. aureus.
Collapse
Affiliation(s)
- Mara J. Campbell
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Karen E. Beenken
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Aura M. Ramirez
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | |
Collapse
|
22
|
Yao L, Huang C, Dai J. Staphylococcus aureus enhances osteoclast differentiation and bone resorption by stimulating the NLRP3 inflammasome pathway. Mol Biol Rep 2023; 50:9395-9403. [PMID: 37817024 DOI: 10.1007/s11033-023-08900-9] [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: 07/28/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Osteomyelitis is one of the most challenging infectious diseases and is mainly caused by Staphylococcus aureus (S. aureus). In this study, we analyzed the effect of S. aureus on osteoclast differentiation and its possible molecular mechanism. METHODS We cultured RAW 264.7 cells with live S. aureus for 5 days. We assessed cell viability and the formation of resorption pits. We tested the NLRP3 inflammasome signaling pathways and measured the mRNA expression levels of osteoclastspecific genes, including TRAP, MMP9, cathepsin K, calcitonin receptor and ATP6V0d2. Furthermore, we analyzed the protein expression levels of the protein in the NF-κB and p38 MAPK signaling pathways to clarify the signaling pathways by which S. aureus promotes osteoclast differentiation. RESULTS Staphylococcus aureus induced NLRP3 inflammasome activation. S. aureus promoted bone resorption and enhanced the expression of osteoclastspecific genes, such as TRAP, MMP9, cathepsin K, calcitonin receptor and ATP6V0d2. MCC950 was used to inhibit NLRP3 inflammasome activity. Osteoclast differentiation and the expression of osteoclastspecific genes induced by S. aureus were inhibited by MCC950 pretreatment. The degradation of IκBα and phosphorylation of P65 were increased under the induction of S. aureus, but proteins in the p38 MAPK signaling pathway did not change significantly. CONCLUSION Staphylococcus aureus induces osteoclast differentiation and promotes bone resorption in vitro, and the NLRP3 inflammasome signaling pathway plays a significant role in this process. S. aureus-induced NLRP3 inflammasome activation was mainly dependent on the NF-κB signaling pathway during osteoclastogenesis.
Collapse
Affiliation(s)
- Ling Yao
- Department of Orthopedic Surgery, The Affiliated Hospital (GROUP) of Putian University, Putian, 351100, Fujian, China
| | - Chongming Huang
- Department of General Surgery, The First People's Hospital of Yibin, No. 65 Wenxing Road, Yibin, 644000, Sichuan, China.
| | - Jiezhi Dai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 YiShan Road, Shanghai, 200230, China.
| |
Collapse
|
23
|
Li X, Pang W, Fan H, Wang H, Zhang L. FRZB affects Staphylococcus aureus‑induced osteomyelitis in human bone marrow derived stem cells by regulating the Wnt/β‑catenin signaling pathway. Exp Ther Med 2023; 26:531. [PMID: 37869648 PMCID: PMC10587868 DOI: 10.3892/etm.2023.12230] [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: 12/06/2022] [Accepted: 03/29/2023] [Indexed: 10/24/2023] Open
Abstract
Osteomyelitis is an infectious disease of bone tissue caused by bacterial infection, which can infect through hematogenous, traumatic or secondary ways and then lead to acute or chronic bone injury and relative clinical symptoms, bringing physical injury and economic burden to patients. Frizzled related protein (FRZB) participates in the regulation of various diseases (osteoarthritis, cardiovascular diseases and types of cancer) by regulating cell proliferation, motility, differentiation and inflammation, while its function in osteomyelitis remains to be elucidated. The present study aimed to uncover the role and underlying mechanism of FRZB mediation in Staphylococcus aureus (S. aureus)-induced osteomyelitis. Human bone marrow derived stem cells (hBMSCs) were treated with S. aureus to imitate an inflammatory osteomyelitis micro-environment in vitro, then mRNA and protein expression were severally assessed by RT-PCR and western blotting. The activity, apoptosis and differentiation of the cells were characterized via CCK-8, caspase-3 activity and Alizarin red sulfate/alkaline phosphatase staining, respectively. Expression levels of FRZB were upregulated in S. aureus-infected hBMSCs. Over-expression of FRZB significantly reduced hBMSC cell viability and differentiation while promoting cell apoptosis with or without S. aureus infection. However, FRZB knockdown reversed these effects. Once Wnt was impeded, the effect of FRZB downregulation was impeded to a great extent. Taken together, FRZB participated to regulate the osteomyelitis by activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xin Li
- Department of Emergency Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550023, P.R. China
| | - Wenyong Pang
- Department of Emergency Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550023, P.R. China
| | - Hongsong Fan
- Department of Emergency Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550023, P.R. China
| | - Hao Wang
- Department of Emergency Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550023, P.R. China
| | - Leibing Zhang
- Department of Emergency Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550023, P.R. China
| |
Collapse
|
24
|
Hu X, Tang J, Yu H, Yang H, Lu X, Zheng D. Preparation of fish collagen and vancomycin microspheres based on microfluidic technology and its application in osteomyelitis. Front Bioeng Biotechnol 2023; 11:1249706. [PMID: 37915548 PMCID: PMC10616836 DOI: 10.3389/fbioe.2023.1249706] [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: 07/09/2023] [Accepted: 08/24/2023] [Indexed: 11/03/2023] Open
Abstract
At present, the clinical treatment of osteomyelitis and osteomyelitis-induced bone defects is challenging, easy to recur, drug toxic side effects, secondary or multiple surgeries, etc. The design of biodegradable composite biomaterials to improve antibiotics in the local precise anti-infection at the same time to complete the repair of bone defects is the current research hot spot. Herein, a composite hydrogel with a double bond at the end (FA-MA) was prepared by affinity addition reaction between fish collagen (FA) and methacrylic anhydride (MA) under photoinitiator initiation conditions, then, FA-MA was amino-activated by EDC/NHC, and vancomycin was attached to FA-MA via amide bonding to prepare FA-MA-Van hydrogels, and finally, the composite hydrogel microspheres were prepared by microfluidic technology. The structure of the hydrogel was confirmed by SEM (elemental analysis), optical microscopy, FTIR, and XPS to confirm the successful preparation. The composite hydrogel microspheres showed the better antimicrobial effect of hydrogel microspheres by bacterial coated plate experiments and SEM morphology results, with the antimicrobial class reaching 99.8%. The results of immunofluorescence staining and X-ray experiments showed that the hydrogel microspheres had a better effect on promoting bone repair. This engineered design of hydrogel microspheres provides clinical significance for treating osteomyelitis at a later stage.
Collapse
Affiliation(s)
- Xiaowu Hu
- Department of Orthopedics, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an Second People’s Hospital, Huaian, Jiangsu, China
| | - Jinshan Tang
- Department of Orthopedics, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an Second People’s Hospital, Huaian, Jiangsu, China
| | - Huaixi Yu
- Department of Orthopedics, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an Second People’s Hospital, Huaian, Jiangsu, China
| | - Hanshi Yang
- Department of Orthopedics, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an Second People’s Hospital, Huaian, Jiangsu, China
| | - Xiaoqing Lu
- Department of Orthopedics, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an Second People’s Hospital, Huaian, Jiangsu, China
| | - Donghui Zheng
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an Second People’s Hospital, Huaian, Jiangsu, China
| |
Collapse
|
25
|
Peilin W, Ying P, Renyuan W, Zhuoxuan L, Zhenwu Y, Mai Z, Jianguo S, Hao Z, Gang Y, Lin L, Haodong L. Size-dependent gold nanoparticles induce macrophage M2 polarization and promote intracellular clearance of Staphylococcus aureus to alleviate tissue infection. Mater Today Bio 2023; 21:100700. [PMID: 37455821 PMCID: PMC10338365 DOI: 10.1016/j.mtbio.2023.100700] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/19/2023] [Accepted: 05/29/2023] [Indexed: 07/18/2023] Open
Abstract
Tissue infection typically results from blood transmission or the direct inoculation of bacteria following trauma. The pathogen-induced destruction of tissue prevents antibiotics from penetrating the infected site, and severe inflammation further impairs the efficacy of conventional treatment. The current study describes the size-dependent induction of macrophage polarization using gold nanoparticles. Gold nanoparticles with a diameter of 50 nm (Au50) can induce M2 polarization in macrophages by inhibiting the NF-κB signaling pathway and stimulate an inflammatory response in the environment by inhibiting the MAPK signaling pathway LPS. Furthermore, the induced polarization and anti-inflammatory effects of the Au50 nanoparticles promoted the osteogenic differentiation of BMSCs in vitro. In addition, the overexpression of TREM2 in macrophage induced by Au50 nanoparticles was found to promote macrophage phagocytosis of Staphylococcus aureus, enhance the fusion of autophagosomes and lysosomes, accelerate the intracellular degradation of S. aureus, in addition to achieving an effective local treatment of osteomyelitis and infectious skin defects in conjunction with inflammatory regulation and accelerating bone regeneration. The findings, therefore, demonstrate that Au50 nanoparticles can be utilized as a promising nanomaterial for in vivo treatment of infections.
Collapse
Affiliation(s)
- Wang Peilin
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Peng Ying
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Wang Renyuan
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Li Zhuoxuan
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Yang Zhenwu
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Zhao Mai
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Song Jianguo
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Zhang Hao
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Yin Gang
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Lin Lin
- School of Chemical and Environmental, Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Lin Haodong
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| |
Collapse
|
26
|
Zeng M, Xu Z, Song ZQ, Li JX, Tang ZW, Xiao S, Wen J. Diagnosis and treatment of chronic osteomyelitis based on nanomaterials. World J Orthop 2023; 14:42-54. [PMID: 36844379 PMCID: PMC9945247 DOI: 10.5312/wjo.v14.i2.42] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/01/2022] [Accepted: 01/17/2023] [Indexed: 02/17/2023] Open
Abstract
Chronic osteomyelitis is a painful and serious disease caused by infected surgical prostheses or infected fractures. Traditional treatment includes surgical debridement followed by prolonged systemic antibiotics. However, excessive antibiotic use has been inducing rapid emergence of antibiotic-resistant bacteria worldwide. Additionally, it is difficult for antibiotics to penetrate internal sites of infection such as bone, thus limiting their efficacy. New approaches to treat chronic osteomyelitis remain a major challenge for orthopedic surgeons. Luckily, the development of nanotechnology has brought new antimicrobial options with high specificity to infection sites, offering a possible way to address these challenges. Substantial progress has been made in constructing antibacterial nanomaterials for treatment of chronic osteomyelitis. Here, we review some current strategies for treatment of chronic osteomyelitis and their underlying mechanisms.
Collapse
Affiliation(s)
- Ming Zeng
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Zheng Xu
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Zhen-Qi Song
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Jie-Xiao Li
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Zhong-Wen Tang
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Sheng Xiao
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Jie Wen
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| |
Collapse
|
27
|
Neutralization of Staphylococcus aureus Protein A Prevents Exacerbated Osteoclast Activity and Bone Loss during Osteomyelitis. Antimicrob Agents Chemother 2023; 67:e0114022. [PMID: 36533935 PMCID: PMC9872667 DOI: 10.1128/aac.01140-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Osteomyelitis caused by Staphylococcus aureus is an important and current health care problem worldwide. Treatment of this infection frequently fails not only due to the increasing incidence of antimicrobial-resistant isolates but also because of the ability of S. aureus to evade the immune system, adapt to the bone microenvironment, and persist within this tissue for decades. We have previously demonstrated the role of staphylococcal protein A (SpA) in the induction of exacerbated osteoclastogenesis and increased bone matrix degradation during osteomyelitis. The aim of this study was to evaluate the potential of using anti-SpA antibodies as an adjunctive therapy to control inflammation and bone damage. By using an experimental in vivo model of osteomyelitis, we demonstrated that the administration of an anti-SpA antibody by the intraperitoneal route prevented excessive inflammatory responses in the bone upon challenge with S. aureus. Ex vivo assays indicated that blocking SpA reduced the priming of osteoclast precursors and their response to RANKL. Moreover, the neutralization of SpA was able to prevent the differentiation and activation of osteoclasts in vivo, leading to reduced expression levels of cathepsin K, reduced expression of markers associated with abnormal bone formation, and decreased trabecular bone loss during osteomyelitis. Taken together, these results demonstrate the feasibility of using anti-SpA antibodies as an antivirulence adjunctive therapy that may prevent the development of pathological conditions that not only damage the bone but also favor bacterial escape from antimicrobials and the immune system.
Collapse
|
28
|
Wang C, Yang Y, Cao Y, Liu K, Shi H, Guo X, Liu W, Hao R, Song H, Zhao R. Nanocarriers for the delivery of antibiotics into cells against intracellular bacterial infection. Biomater Sci 2023; 11:432-444. [PMID: 36503914 DOI: 10.1039/d2bm01489k] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The barrier function of host cells enables intracellular bacteria to evade the lethality of the host immune system and antibiotics, thereby causing chronic and recurrent infections that seriously threaten human health. Currently, the main clinical strategy for the treatment of intracellular bacterial infections involves the use of long-term and high-dose antibiotics. However, insufficient intracellular delivery of antibiotics along with various resistance mechanisms not only weakens the efficacy of current therapies but also causes serious adverse drug reactions, further increasing the disease and economic burden. Improving the delivery efficiency, intracellular accumulation, and action time of antibiotics remains the most economical and effective way to treat intracellular bacterial infections. The rapid development of nanotechnology provides a strategy to efficiently deliver antibiotics against intracellular bacterial infections into cells. In this review, we summarize the types of common intracellular pathogens, the difficulties faced by antibiotics in the treatment of intracellular bacterial infections, and the research progress of several types of representative nanocarriers for the delivery of antibiotics against intracellular bacterial infections that have emerged in recent years. This review is expected to provide a reference for further elucidating the intracellular transport mechanism of nanocarrier-drug complexes, designing safer and more effective nanocarriers and establishing new strategies against intracellular bacterial infection.
Collapse
Affiliation(s)
- Chao Wang
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Yi Yang
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Yuanyuan Cao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Kaixin Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Hua Shi
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Xudong Guo
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Wanying Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Rongzhang Hao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Hongbin Song
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Rongtao Zhao
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| |
Collapse
|
29
|
Hexapeptide decorated β-cyclodextrin delivery system for targeted therapy of bone infection. J Control Release 2023; 353:337-349. [PMID: 36462641 DOI: 10.1016/j.jconrel.2022.11.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Successfully treating bone infections is a major orthopedic challenge. Clinically, oral, intravenous, or intramuscular injections of drugs are usually used for direct or complementary treatment. However, once the drug enters the system, it circulates throughout the body, leading to an insufficient local dose and limiting the therapeutic effect because of the lack of targeting in the drug system. In this study, β-cyclodextrin, modified with poly (ethylene glycol) [PEG] and aspartic acid hexapeptide (Asp6-β-CD), was used to specifically target the hydroxyapatite (HA) component of the bone. It was then loaded with norfloxacin (NFX) to treat bone infections. The antibacterial ability of NFX was enhanced by loading it into Asp6-β-CD, because the solubility of Asp6-β-CD@NFX increased significantly. Moreover, Asp6-β-CD could target bone tissue in nude mice and showed significantly enhanced accumulation (10 times) than the unmodified β-CD. In addition, in a rat model of osteomyelitis, Asp6-β-CD@NFX targeted HA well and exerted its antibacterial activity, which reduced inflammation and promoted bone tissue repair. This study indicates that the Asp6-β-CD based drug delivery system can efficiently target bone tissue to enable potential applications for treating bone-related diseases.
Collapse
|
30
|
Heng BC, Bai Y, Li X, Lim LW, Li W, Ge Z, Zhang X, Deng X. Electroactive Biomaterials for Facilitating Bone Defect Repair under Pathological Conditions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204502. [PMID: 36453574 PMCID: PMC9839869 DOI: 10.1002/advs.202204502] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/24/2022] [Indexed: 06/02/2023]
Abstract
Bone degeneration associated with various diseases is increasing due to rapid aging, sedentary lifestyles, and unhealthy diets. Living bone tissue has bioelectric properties critical to bone remodeling, and bone degeneration under various pathological conditions results in significant changes to these bioelectric properties. There is growing interest in utilizing biomimetic electroactive biomaterials that recapitulate the natural electrophysiological microenvironment of healthy bone tissue to promote bone repair. This review first summarizes the etiology of degenerative bone conditions associated with various diseases such as type II diabetes, osteoporosis, periodontitis, osteoarthritis, rheumatoid arthritis, osteomyelitis, and metastatic osteolysis. Next, the diverse array of natural and synthetic electroactive biomaterials with therapeutic potential are discussed. Putative mechanistic pathways by which electroactive biomaterials can mitigate bone degeneration are critically examined, including the enhancement of osteogenesis and angiogenesis, suppression of inflammation and osteoclastogenesis, as well as their anti-bacterial effects. Finally, the limited research on utilization of electroactive biomaterials in the treatment of bone degeneration associated with the aforementioned diseases are examined. Previous studies have mostly focused on using electroactive biomaterials to treat bone traumatic injuries. It is hoped that this review will encourage more research efforts on the use of electroactive biomaterials for treating degenerative bone conditions.
Collapse
Affiliation(s)
- Boon Chin Heng
- Central LaboratoryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- School of Medical and Life SciencesSunway UniversityDarul EhsanSelangor47500Malaysia
| | - Yunyang Bai
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xiaochan Li
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Lee Wei Lim
- Neuromodulation LaboratorySchool of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongPokfulamHong KongP. R. China
| | - Wang Li
- Department of Biomedical EngineeringPeking UniversityBeijing100871P. R. China
| | - Zigang Ge
- Department of Biomedical EngineeringPeking UniversityBeijing100871P. R. China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical DevicesNMPA Key Laboratory for Dental MaterialsBeijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xuliang Deng
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical DevicesNMPA Key Laboratory for Dental MaterialsBeijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| |
Collapse
|
31
|
Li Y, Chen L, Lin M, Wang C, Zhang R, Li Y, Zou Q. Micro-CT analysis of osteomyelitis of rabbit tibial for model establishment and biomaterials application in tissue engineering. Heliyon 2022; 8:e12471. [PMID: 36643303 PMCID: PMC9834739 DOI: 10.1016/j.heliyon.2022.e12471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 07/08/2022] [Accepted: 12/12/2022] [Indexed: 12/25/2022] Open
Abstract
Osteomyelitis is one of the most difficult diseases to treat in orthopedics field. The construction of animal models of osteomyelitis is now more mature but still lacks a deeper criterion other than "successful infection". In this work, rabbit tibial osteomyelitis model with S. aureus infection was established. Whole tibia, cortical bone around bone window, and tibial condyle were characterized in considerable detail using micro-CT and other means at 2/4/6 weeks, respectively. The results show that in addition to the obvious inflammatory response and bone destruction, osteomyelitis caused some other effects on compact and cancellous bone, and in particular, changes in bone mineral density after infection were of interest. Although the modeling groups all exhibited osteolysis and bone loss, their overall bone mineral density averages and those of the control groups were mostly in the range of 870 mg/cm3 to 920 mg/cm3, without statistical difference. The results suggest that overall bone mineral density is determined by both bone destruction conditions and the amount of dead bone deposition. This work provides a reference basis for the selection of time points for the subsequent animal models establishment and some valuable reference indicators of the application of biomaterials in tissue engineering.
Collapse
Affiliation(s)
- Yufan Li
- Analytical and Testing Center, Nano Biomaterials Research Center, Sichuan University, No. 29 Jiuyanqiao Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Li Chen
- Analytical and Testing Center, Sichuan University, No. 29 Jiuyanqiao Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Mingyue Lin
- Analytical and Testing Center, Nano Biomaterials Research Center, Sichuan University, No. 29 Jiuyanqiao Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Chenxin Wang
- Analytical and Testing Center, Nano Biomaterials Research Center, Sichuan University, No. 29 Jiuyanqiao Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Rui Zhang
- Analytical and Testing Center, Nano Biomaterials Research Center, Sichuan University, No. 29 Jiuyanqiao Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Yubao Li
- Analytical and Testing Center, Nano Biomaterials Research Center, Sichuan University, No. 29 Jiuyanqiao Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Qin Zou
- Analytical and Testing Center, Nano Biomaterials Research Center, Sichuan University, No. 29 Jiuyanqiao Wangjiang Road, Chengdu, Sichuan, 610064, China
| |
Collapse
|
32
|
Moradi M, Mohabatkar H, Behbahani M, Dini G. Application of G-quadruplex aptamer conjugated MSNs to deliver ampicillin for suppressing S. aureus biofilm on mice bone. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
33
|
Chen J, Wang X, Li J, Zhang H, Li Y, Deng P, Feng W, Qi X, Ye P, Li J, Zeng J, Zeng Y, Li J, Xie S. Research Status and Hotspots of Chronic Osteomyelitis: A Bibliometric and Visualized Analysis. Orthop Surg 2022; 14:3378-3389. [PMID: 36266919 PMCID: PMC9732618 DOI: 10.1111/os.13512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 07/26/2022] [Accepted: 08/23/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE The treatment of chronic osteomyelitis (COM) is extremely challenging for physicians and patients. It is of great significance to explore the research status, development trend and future research hotspots in the field of COM to promote the development of this field. This study is aimed to explore the global research status of COM and predict its future research hotspots based on bibliometric and visualized analysis. METHODS Web of Science core collection database was used to search the related literature of COM from 1994 to 2020. All data were imported into Microsoft Excel 2019 for collation. Additionally, the literature quality of countries, authors, journals, and institutions is evaluated. The VOS viewer software was used for conducting co-analysis, co-citation analysis, and keyword co-occurrence analysis of literature to analyze the global status and predict the future hotspots of the COM field. RESULTS A total of 726 articles were retrieved in this study. The number of global publications shows a trend of wave growth, but the increase is not significant. It is expected that the number of COM articles will remain at more than 50 per year in the next decade. The COM literature published in the United States (Publications = 160, H index = 37, average citations per item = 28.63) is of the highest quality. Girschick HJ (Publications = 16, H index = 14, average citations per item = 52.25) is the most contributed scholar in the field of COM. UNIV IOWA (Publications = 15, H index = 11, average citations per item = 57.27) and UNIV WURZBURG (Publications = 18, H index = 15, average citations per item = 47.5) are influential institutions in the field of COM. The results of co-occurrence analysis show that the field of COM can be roughly divided into the following five modules: COM surgical research, COM basic research, COM diagnosis-related research, chronic recurrent multifocal osteomyelitis (CRMO)-related research, risk factors of COM. Risk factors of COM are the module with the highest concentration of hot words. CONCLUSION COM-related research will continue to develop further in the next decade. The diagnosis research and risk factors of COM are the most popular research modules in recent years. Some controversial or troubled issues including the efficacy of perforator flap and fascia flap covering soft tissue, searching exclusive detection methods for the diagnosis of COM and bisphosphonates and biological agents in the treatment of CRMO may lead to the development of the COM field.
Collapse
Affiliation(s)
- Jinlun Chen
- The Three Department of OrthopedicsThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina,The First Clinical Medical CollegeGuangzhou University of Chinese MedicineGuangzhouChina
| | - Xingyu Wang
- The First Department of OrthopedicsJiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Jin Li
- The Fourth Clinical Medical CollegeGuangzhou University of Chinese MedicineShenzhenChina
| | - Haitao Zhang
- The First Clinical Medical CollegeGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yijin Li
- The First Clinical Medical CollegeGuangzhou University of Chinese MedicineGuangzhouChina
| | - Peng Deng
- The Three Department of OrthopedicsThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Wenjun Feng
- The Three Department of OrthopedicsThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Xinyu Qi
- The Three Department of OrthopedicsThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Pengcheng Ye
- The Three Department of OrthopedicsThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Jiahao Li
- The First Clinical Medical CollegeGuangzhou University of Chinese MedicineGuangzhouChina
| | - Jianchun Zeng
- The Three Department of OrthopedicsThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Yirong Zeng
- The Three Department of OrthopedicsThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Jie Li
- The Three Department of OrthopedicsThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Shuihua Xie
- The First Department of OrthopedicsJiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| |
Collapse
|
34
|
Ito Y, Amagai M. Controlling skin microbiome as a new bacteriotherapy for inflammatory skin diseases. Inflamm Regen 2022; 42:26. [PMID: 36045395 PMCID: PMC9434865 DOI: 10.1186/s41232-022-00212-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/10/2022] [Indexed: 11/12/2022] Open
Abstract
The skin serves as the interface between the human body and the environment and interacts with the microbial community. The skin microbiota consists of microorganisms, such as bacteria, fungi, mites, and viruses, and they fluctuate depending on the microenvironment defined by anatomical location and physiological function. The balance of interactions between the host and microbiota plays a pivotal role in the orchestration of skin homeostasis; however, the disturbance of the balance due to an alteration in the microbial communities, namely, dysbiosis, leads to various skin disorders. Recent developments in sequencing technology have provided new insights into the structure and function of skin microbial communities. Based on high-throughput sequencing analysis, a growing body of evidence indicates that a new treatment using live bacteria, termed bacteriotherapy, is a feasible therapeutic option for cutaneous diseases caused by dysbiosis. In particular, the administration of specific bacterial strains has been investigated as an exclusionary treatment strategy against pathogens associated with chronic skin disorders, whereas the safety, efficacy, and sustainability of this therapeutic approach using isolated live bacteria need to be further explored. In this review, we summarize our current understanding of the skin microbiota, as well as therapeutic strategies using characterized strains of live bacteria for skin inflammatory diseases. The ecosystem formed by interactions between the host and skin microbial consortium is still largely unexplored; however, advances in our understanding of the function of the skin microbiota at the strain level will lead to the development of new therapeutic methods.
Collapse
Affiliation(s)
- Yoshihiro Ito
- Department of Dermatology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| |
Collapse
|
35
|
Dehority W, Morley VJ, Domman DB, Daly SM, Triplett KD, Disch K, Varjabedian R, Yousey A, Mortaji P, Hill D, Oyebamiji O, Guo Y, Schwalm K, Hall PR, Dinwiddie D, Femling J. Genomic characterization of Staphylococcus aureus isolates causing osteoarticular infections in otherwise healthy children. PLoS One 2022; 17:e0272425. [PMID: 36037235 PMCID: PMC9423648 DOI: 10.1371/journal.pone.0272425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 07/19/2022] [Indexed: 12/03/2022] Open
Abstract
Background Pediatric osteoarticular infections are commonly caused by Staphylococcus aureus. The contribution of S. aureus genomic variability to pathogenesis of these infections is poorly described. Methods We prospectively enrolled 47 children over 3 1/2 years from whom S. aureus was isolated on culture—12 uninfected with skin colonization, 16 with skin abscesses, 19 with osteoarticular infections (four with septic arthritis, three with acute osteomyelitis, six with acute osteomyelitis and septic arthritis and six with chronic osteomyelitis). Isolates underwent whole genome sequencing, with assessment for 254 virulence genes and any mutations as well as creation of a phylogenetic tree. Finally, isolates were compared for their ability to form static biofilms and compared to the genetic analysis. Results No sequence types predominated amongst osteoarticular infections. Only genes involved in evasion of host immune defenses were more frequently carried by isolates from osteoarticular infections than from skin colonization (p = .02). Virulence gene mutations were only noted in 14 genes (three regulating biofilm formation) when comparing isolates from subjects with osteoarticular infections and those with skin colonization. Biofilm results demonstrated large heterogeneity in the isolates’ capacity to form static biofilms, with healthy control isolates producing more robust biofilm formation. Conclusions S. aureus causing osteoarticular infections are genetically heterogeneous, and more frequently harbor genes involved in immune evasion than less invasive isolates. However, virulence gene carriage overall is similar with infrequent mutations, suggesting that pathogenesis of S. aureus osteoarticular infections may be primarily regulated at transcriptional and/or translational levels.
Collapse
Affiliation(s)
- Walter Dehority
- Department of Pediatrics, The University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
- * E-mail:
| | - Valerie J. Morley
- Department of Internal Medicine, The University of New Mexico School of Medicine, Center for Global Health, Albuquerque, New Mexico, United States of America
| | - Daryl B. Domman
- Department of Internal Medicine, The University of New Mexico School of Medicine, Center for Global Health, Albuquerque, New Mexico, United States of America
| | - Seth M. Daly
- Department of Pharmaceutical Sciences, The University of New Mexico College of Pharmacy, Albuquerque, New Mexico, United States of America
| | - Kathleen D. Triplett
- Department of Pharmaceutical Sciences, The University of New Mexico College of Pharmacy, Albuquerque, New Mexico, United States of America
| | - Kylie Disch
- Department of Pediatrics, The University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | | | - Aimee Yousey
- Department of Emergency Medicine, The University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | - Parisa Mortaji
- Department of Internal Medicine, The University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Deirdre Hill
- The University of New Mexico Clinical and Translational Science Center, Albuquerque, New Mexico, United States of America
| | - Olufunmilola Oyebamiji
- Division of Molecular Medicine, The University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Yan Guo
- Division of Molecular Medicine, The University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Kurt Schwalm
- Department of Pediatrics, The University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | - Pamela R. Hall
- Department of Pharmaceutical Sciences, The University of New Mexico College of Pharmacy, Albuquerque, New Mexico, United States of America
| | - Darrell Dinwiddie
- Department of Pediatrics, The University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| | - Jon Femling
- Department of Emergency Medicine, The University of New Mexico School of Medicine, Albuquerque, New Mexico, United States of America
| |
Collapse
|
36
|
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
|
37
|
Karau M, Schmidt-Malan S, Mandrekar J, Lehoux D, Schuch R, Cassino C, Patel R. Locally delivered antistaphylococcal lysin exebacase or CF-296 is active in methicillin-resistant Staphylococcus aureus implant-associated osteomyelitis. J Bone Jt Infect 2022; 7:169-175. [PMID: 36032801 PMCID: PMC9399932 DOI: 10.5194/jbji-7-169-2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/07/2022] [Indexed: 11/11/2022] Open
Abstract
Abstract. Introduction: Staphylococcus aureus is the most common cause of orthopedic infections and can be
challenging to treat, especially in the presence of a foreign body. The
antistaphylococcal lysins exebacase and CF-296 have rapid bactericidal
activity, a low propensity for resistance development, and synergize with
some antibiotics.
Methods: Rabbit implant-associated osteomyelitis was induced by drilling
into the medial tibia followed by locally delivering exebacase, CF-296, or
lysin carrier. A titanium screw colonized with methicillin-resistant S. aureus (MRSA) IDRL-6169 was inserted.
Intravenous daptomycin or saline was administered and continued daily for
4 d. On day 5, rabbits were euthanized, and the tibiae and implants were
collected for culture. Results were reported as log10 colony forming units (cfu) per gram of bone or log10 cfu per implant, and comparisons among the six groups were performed using the
Wilcoxon rank sum test.
Results: Based on implant and bone cultures, all treatments resulted in
significantly lower bacterial counts than those of controls (P≤0.0025).
Exebacase alone or with daptomycin as well as CF-296 with daptomycin were more
active than daptomycin alone (P≤0.0098) or CF-296 alone (P≤0.0154)
based on implant cultures. CF-296 with daptomycin was more active than
either CF-296 alone (P=0.0040) or daptomycin alone (P=0.0098) based on
bone cultures.
Conclusion: Local delivery of either exebacase or CF-296 offers a promising
complement to conventional antibiotics in implant-associated infections.
Collapse
Affiliation(s)
- Melissa Karau
- Division of Clinical Microbiology, Department of Laboratory Medicine
and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Suzannah Schmidt-Malan
- Division of Clinical Microbiology, Department of Laboratory Medicine
and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Jay Mandrekar
- Division of Biomedical Statistics and Informatics, Department of
Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine
and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Public Health, Infectious Diseases, and Occupational
Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
38
|
Shi X, Wu Y, Ni H, Li M, Zhang C, Qi B, Wei M, Wang T, Xu Y. Antibiotic-loaded calcium sulfate in clinical treatment of chronic osteomyelitis: a systematic review and meta-analysis. J Orthop Surg Res 2022; 17:104. [PMID: 35183215 PMCID: PMC8858512 DOI: 10.1186/s13018-022-02980-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/27/2022] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Present work was aimed to gather accessible evidence on the eradication rates and related postoperative complications of antibiotic-loaded calcium sulfate (CS) as an implant in the treatment of chronic osteomyelitis (COM).
Methods
Databases including PubMed, EMBASE, Medline, Ovid and Cochrane library were searched from their dates of initiation until November 2021. Two independent authors scrutinized the relevant studies based on the effectiveness of radical debridement combined with antibiotic-loaded CS for COM; data extraction and quality assessment of the Methodological Index for Non-Randomized Studies (MINORS) criteria were also performed by the authors. In addition, clinical efficacy mainly depended on the evaluation of eradication rates and complications, and all the extracted data are pooled and analyzed by STATA 16.0.
Results
A total of 16 studies with 917 patients (920 locations) were recruited, with an overall eradication rate of 92%. Moreover, the overall reoperation rate, overall refracture rate, overall delayed wound healing rate, and the rate of aseptic wound leakage were 9.0%, 2.0%, 20.0%, and 12.0%, respectively. Moreover, the choice of tobramycin-loaded CS or vancomycin combined with gentamicin-loaded CS did not affect the eradication rate, and the incidence of postoperative complications in COM patients (all $$P>0.05$$
P
>
0.05
). The general quality of the included studies was fair.
Conclusions
Our meta-analysis indicated that the overall eradication rate of COM treated with antibiotic-loaded CS was 92%. Delayed healing is the most common postoperative complication. The choice of tobramycin-loaded CS or vancomycin combined with gentamicin-loaded CS did not affect the eradication rate and the incidence of postoperative complications in COM patients.
Collapse
|
39
|
Fallah F, Shirani K, Khorvash F, Soltani R, Ataie B, Tarrahi M. The effect of Vitamin B 6 in the prevention of hematological adverse effects of linezolid in patients with chronic osteomyelitis: A randomized double-blind placebo-controlled clinical trial. Adv Biomed Res 2022; 11:67. [PMID: 36325173 PMCID: PMC9621342 DOI: 10.4103/abr.abr_274_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 11/06/2022] Open
Abstract
Background: Due to the contradictory results of the effects of Vitamin B6 in reducing the hematotoxic effects of linezolid, the present study aimed to investigate the possible role of Vitamin B6 administration in reducing linezolid-related hematological toxicities in patients with chronic osteomyelitis. Materials and Methods: In a randomized double-blind placebo-controlled clinical trial, patients with chronic osteomyelitis were randomly divided into two groups (n = 40 each): the intervention group received Vitamin B640 mg twice daily from the beginning of treatment with linezolid and the control group received placebo with linezolid, both for 21 days. Blood variables including hemoglobin (Hb), white blood cells (WBC), and platelets (PLT) were measured at baseline and at the end of the 1st, 2nd, and 3rd weeks (days 7, 14, and 21) of the intervention. Results: There was no significant difference between the groups regarding the count of WBC and PLT and level of Hb at evaluated time points. Furthermore, there was a significant decreasing trend in all parameters within both groups; however, the decreasing trend of both PLT and WBC was slower in the intervention (Vitamin B6) group compared to the placebo group. Conclusion: Vitamin B6 has no significant effect in the reduction of hematological adverse effects of linezolid in chronic osteomyelitis patients. However, it could retard the decreasing trend of WBC and PLT counts.
Collapse
|
40
|
Osteogenic and anti-inflammatory potential of oligochitosan nanoparticles in treating osteomyelitis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 135:112681. [DOI: 10.1016/j.msec.2022.112681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 01/10/2022] [Accepted: 01/21/2022] [Indexed: 12/12/2022]
|
41
|
Activity of Omadacycline in Rat Methicillin-Resistant Staphylococcus aureus Osteomyelitis. Antimicrob Agents Chemother 2021; 66:e0170321. [PMID: 34723626 PMCID: PMC8765317 DOI: 10.1128/aac.01703-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Omadacycline, vancomycin, and rifampin, as well as rifampin combination therapies, were evaluated in an experimental rat model of methicillin-resistant Staphylococcus aureus (MRSA) osteomyelitis. All treatment groups had less MRSA recovered than saline-treated animals. The emergence of rifampin resistance was observed in 3 of 16 animals with rifampin monotherapy and none with rifampin combination therapy. After treatment, the median tibial bacterial loads were 6.04, 0.1, 4.81, and 5.24 log10 CFU/g for saline-, rifampin-, vancomycin-, and omadacycline-treated animals, respectively. Omadacycline or vancomycin administered with rifampin yielded no detectable MRSA. Omadacycline administered with rifampin deserves evaluation in humans as a potential treatment for osteomyelitis.
Collapse
|
42
|
Carvalho SM, Freitas CS, Oliveira AS, Saraiva LM. Staphylococcus aureus flavohaemoglobin contributes to early stage biofilm development under nitrosative stress. FEMS Microbiol Lett 2021; 368:6402901. [PMID: 34665259 DOI: 10.1093/femsle/fnab131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is a Gram-positive bacterium with capacity to form biofilms, which constitute an important resistance mechanism and virulence factor. Flavohaemoglobin (Hmp) is a major nitric oxide (NO) detoxifier of several bacteria, including S. aureus. Although Hmp has a well-known physiological role linked to response of planktonic cells to nitrosative stress, its contribution to biofilm formation remains unaddressed. Hence, in this work, we investigated the role of Hmp in biofilm development of a methicillin-resistant S. aureus strain. For this purpose, we exposed the hmp mutant to nitrosative stress and examined its behaviour along biofilm development. We observed that cells inactivated in hmp and grown under nitrosative stress conditions have significantly impaired capacity to develop early stage biofilms. Furthermore, the wild-type biofilm phenotype was fully restored by trans-complementation of hmp in the hmp mutant. Coculture studies of NO-producing macrophages with S. aureus revealed that the hmp mutant has significantly lower capacity to develop biofilm biomass when compared with the wild type. Thus, we concluded that the pathogen S. aureus relies on Hmp to establish viable biofilms in the presence of cells of the host innate immune system.
Collapse
Affiliation(s)
- Sandra M Carvalho
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Cláudia S Freitas
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Ana S Oliveira
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Lígia M Saraiva
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| |
Collapse
|
43
|
Infections @ Trauma/Orthopedic Implants: Recent Advances on Materials, Methods, and Microbes-A Mini-Review. MATERIALS 2021; 14:ma14195834. [PMID: 34640231 PMCID: PMC8510481 DOI: 10.3390/ma14195834] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 12/22/2022]
Abstract
Implants and materials are indispensable in trauma and orthopedic surgery. The continuous improvements of implant design have resulted in an optimized mechanical function that supports tissue healing and restoration of function. One of the still unsolved problems with using implants and materials is infection. Trauma and material implantation change the local inflammatory situation and enable bacterial survival and material colonization. The main pathogen in orthopedic infections is Staphylococcus aureus. The research efforts to optimize antimicrobial surfaces and to develop new anti-infective strategies are enormous. This mini-review focuses on the publications from 2021 with the keywords S. aureus AND (surface modification OR drug delivery) AND (orthopedics OR trauma) AND (implants OR nails OR devices). The PubMed search yielded 16 original publications and two reviews. The original papers reported the development and testing of anti-infective surfaces and materials: five studies described an implant surface modification, three developed an implant coating for local antibiotic release, the combination of both is reported in three papers, while five publications are on antibacterial materials but not metallic implants. One review is a systematic review on the prevention of stainless-steel implant-associated infections, the other addressed the possibilities of mixed oxide nanotubes. The complexity of the approaches differs and six of them showed efficacy in animal studies.
Collapse
|
44
|
Van Belkum A, Gros MF, Ferry T, Lustig S, Laurent F, Durand G, Jay C, Rochas O, Ginocchio CC. Novel strategies to diagnose prosthetic or native bone and joint infections. Expert Rev Anti Infect Ther 2021; 20:391-405. [PMID: 34384319 DOI: 10.1080/14787210.2021.1967745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Bone and Joint Infections (BJI) are medically important, costly and occur in native and prosthetic joints. Arthroplasties will increase significantly in absolute numbers over time as well as the incidence of Prosthetic Joint Infections (PJI). Diagnosis of BJI and PJI is sub-optimal. The available diagnostic tests have variable effectiveness, are often below standard in sensitivity and/or specificity, and carry significant contamination risks during the collection of clinical samples. Improvement of diagnostics is urgently needed. AREAS COVERED We provide a narrative review on current and future diagnostic microbiology technologies. Pathogen identification, antibiotic resistance detection, and assessment of the epidemiology of infections via bacterial typing are considered useful for improved patient management. We confirm the continuing importance of culture methods and successful introduction of molecular, mass spectrometry-mediated and next-generation genome sequencing technologies. The diagnostic algorithms for BJI must be better defined, especially in the context of diversity of both disease phenotypes and clinical specimens rendered available. EXPERT OPINION Whether interventions in BJI or PJI are surgical or chemo-therapeutic (antibiotics and bacteriophages included), prior sensitive and specific pathogen detection remains a therapy-substantiating necessity. Innovative tests for earlier and more sensitive and specific detection of bacterial pathogens in BJI are urgently needed.
Collapse
Affiliation(s)
- Alex Van Belkum
- bioMérieux, Open Innovation and Partnerships, 3 Route De Port Michaud, La Balme Les Grottes, France
| | | | - Tristan Ferry
- Service Des Maladies Infectieuses Et Tropicales, Hospices Civils De Lyon, Hôpital De La Croix-Rousse, Lyon, France.,Maladies Infectieuses, Université Claude Bernard Lyon 1, Villeurbanne, France.,Centre Interrégional De Référence Pour La Prise En Charge Des Infections Ostéo-articulaires Complexes (Crioac Lyon), Hôpital De La Croix-Rousse, Lyon, France.,Ciri - Centre International De Recherche En Infectiologie, Inserm, U1111, Université́ Claude Bernard Lyon 1CNRS, UMR5308, Ecole Normale Supérieure De Lyon, Univ Lyon, Lyon, France
| | - Sebastien Lustig
- Maladies Infectieuses, Université Claude Bernard Lyon 1, Villeurbanne, France.,Service De Chirurgie Orthopédique, Hôpital De La Croix-Rousse, Lyon, France
| | - Frédéric Laurent
- Service Des Maladies Infectieuses Et Tropicales, Hospices Civils De Lyon, Hôpital De La Croix-Rousse, Lyon, France.,Ciri - Centre International De Recherche En Infectiologie, Inserm, U1111, Université́ Claude Bernard Lyon 1CNRS, UMR5308, Ecole Normale Supérieure De Lyon, Univ Lyon, Lyon, France
| | | | - Corinne Jay
- bioMérieux, BioFire Development Emea, Grenoble, France
| | - Olivier Rochas
- Corporate Business Development, bioMérieux, Marcy-l'Étoile, France
| | | |
Collapse
|
45
|
Nasser A, Dallal MMS, Jahanbakhshi S, Azimi T, Nikouei L. Staphylococcus aureus: biofilm formation and strategies against it. Curr Pharm Biotechnol 2021; 23:664-678. [PMID: 34238148 DOI: 10.2174/1389201022666210708171123] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/09/2021] [Accepted: 05/31/2021] [Indexed: 11/22/2022]
Abstract
The formation of Staphylococcus aureus biofilm causes significant infections in the human body. Biofilm forms through the aggregation of bacterial species and brings about many complications. It mediates drug resistance and persistence and facilitates the recurrence of infection at the end of antimicrobial therapy. Biofilm formation goes through a series of steps to complete, and any interference in these steps can disrupt its formation. Such interference may occur at any stage of biofilm production, including attachment, monolayer formation, and accumulation. Interfering agents can act as quorum sensing inhibitors and interfere in the functionality of quorum sensing receptors, attachment inhibitors and affect the cell hydrophobicity. Among these inhibiting strategies, attachment inhibitors could serve as the best agents against biofilm formation. If pathogens abort the attachment, the following stages of biofilm formation, e.g., accumulation and dispersion, will fail to materialize. Inhibition at this stage leads to suppression of virulence factors and invasion. One of the best-known inhibitors is a chelator that collects metal, Fe+, Zn+, and magnesium critical for biofilm formation. These influential factors in the binding and formation of biofilm are investigated, and the coping strategy is discussed. This review examines the stages of biofilm formation and determines what factors interfere in the continuity of these steps. Finally, the inhibition strategies are investigated, reviewed, and discussed. Keywords: Biofilm, Staphylococcus, Biofilm inhibitor, Dispersion, Antibiofilm agent, EPS, PIA.
Collapse
Affiliation(s)
- Ahmad Nasser
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Shiva Jahanbakhshi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Taher Azimi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Nikouei
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Parente R, Possetti V, Schiavone ML, Campodoni E, Menale C, Loppini M, Doni A, Bottazzi B, Mantovani A, Sandri M, Tampieri A, Sobacchi C, Inforzato A. 3D Cocultures of Osteoblasts and Staphylococcus aureus on Biomimetic Bone Scaffolds as a Tool to Investigate the Host-Pathogen Interface in Osteomyelitis. Pathogens 2021; 10:pathogens10070837. [PMID: 34357987 PMCID: PMC8308613 DOI: 10.3390/pathogens10070837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022] Open
Abstract
Osteomyelitis (OM) is an infectious disease of the bone primarily caused by the opportunistic pathogen Staphylococcus aureus (SA). This Gram-positive bacterium has evolved a number of strategies to evade the immune response and subvert bone homeostasis, yet the underlying mechanisms remain poorly understood. OM has been modeled in vitro to challenge pathogenetic hypotheses in controlled conditions, thus providing guidance and support to animal experimentation. In this regard, traditional 2D models of OM inherently lack the spatial complexity of bone architecture. Three-dimensional models of the disease overcome this limitation; however, they poorly reproduce composition and texture of the natural bone. Here, we developed a new 3D model of OM based on cocultures of SA and murine osteoblastic MC3T3-E1 cells on magnesium-doped hydroxyapatite/collagen I (MgHA/Col) scaffolds that closely recapitulate the bone extracellular matrix. In this model, matrix-dependent effects were observed in proliferation, gene transcription, protein expression, and cell–matrix interactions both of the osteoblastic cell line and of bacterium. Additionally, these had distinct metabolic and gene expression profiles, compared to conventional 2D settings, when grown on MgHA/Col scaffolds in separate monocultures. Our study points to MgHA/Col scaffolds as biocompatible and bioactive matrices and provides a novel and close-to-physiology tool to address the pathogenetic mechanisms of OM at the host–pathogen interface.
Collapse
Affiliation(s)
- Raffaella Parente
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
| | - Valentina Possetti
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
| | - Maria Lucia Schiavone
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
- National Research Council-Institute for Genetic and Biomedical Research (CNR-IRGB), Milan Unit, 20089 Rozzano, Italy;
| | - Elisabetta Campodoni
- National Research Council-Institute of Science and Technology for Ceramics (CNR-ISTEC), 48018 Faenza, Italy; (E.C.); (M.S.); (A.T.)
| | - Ciro Menale
- National Research Council-Institute for Genetic and Biomedical Research (CNR-IRGB), Milan Unit, 20089 Rozzano, Italy;
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
| | - Mattia Loppini
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
| | - Andrea Doni
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
| | - Barbara Bottazzi
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
| | - Alberto Mantovani
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
- The William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK
| | - Monica Sandri
- National Research Council-Institute of Science and Technology for Ceramics (CNR-ISTEC), 48018 Faenza, Italy; (E.C.); (M.S.); (A.T.)
| | - Anna Tampieri
- National Research Council-Institute of Science and Technology for Ceramics (CNR-ISTEC), 48018 Faenza, Italy; (E.C.); (M.S.); (A.T.)
- National Research Council-Institute of Nanostructured Material (CNR-ISMN), 40129 Bologna, Italy
| | - Cristina Sobacchi
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
- National Research Council-Institute for Genetic and Biomedical Research (CNR-IRGB), Milan Unit, 20089 Rozzano, Italy;
- Correspondence: (C.S.); (A.I.); Tel.: +39-028-224-5153 (C.S.); +39-028-224-5132 (A.I.)
| | - Antonio Inforzato
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (R.P.); (V.P.); (M.L.S.); (M.L.); (A.D.); (B.B.); (A.M.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
- Correspondence: (C.S.); (A.I.); Tel.: +39-028-224-5153 (C.S.); +39-028-224-5132 (A.I.)
| |
Collapse
|
47
|
Zhang F, Wang B, Liu S, Chen Y, Lin Y, Liu Z, Zhang X, Yu B. Bacillus subtilis revives conventional antibiotics against Staphylococcus aureus osteomyelitis. Microb Cell Fact 2021; 20:102. [PMID: 34001083 PMCID: PMC8130150 DOI: 10.1186/s12934-021-01592-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/08/2021] [Indexed: 11/28/2022] Open
Abstract
As treatment of Staphylococcus aureus (S. aureus) osteomyelitis is often hindered by the development of antibiotic tolerance, novel antibacterial therapeutics are required. Here we found that the cell-free supernatant of Bacillus subtilis (B. subtilis CFS) killed planktonic and biofilm S. aureus, and increased S. aureus susceptibility to penicillin and gentamicin as well. Further study showed that B. subtilis CFS suppressed the expression of the genes involved in adhesive molecules (Cna and ClfA), virulence factor Hla, quorum sensing (argA, argB and RNAIII) and biofilm formation (Ica and sarA) in S. aureus. Additionally, our data showed that B. subtilis CFS changed the membrane components and increased membrane permeabilization of S. aureus. Finally, we demonstrated that B. subtilis CFS increased considerably the susceptibility of S. aureus to penicillin and effectively reduced S. aureus burdens in a mouse model of implant-associated osteomyelitis. These findings support that B. subtilis CFS may be a potential resistance-modifying agent for β-lactam antibiotics against S. aureus.
Collapse
Affiliation(s)
- Fan Zhang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bowei Wang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Shiluan Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuhui Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yihuang Lin
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zixian Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xianrong Zhang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong, China. .,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong, China. .,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
48
|
Xiao H, Xu Y. Overexpression of Apolipoprotein C1 (APOC1) in Clear Cell Renal Cell Carcinoma and Its Prognostic Significance. Med Sci Monit 2021; 27:e929347. [PMID: 33591959 PMCID: PMC7896428 DOI: 10.12659/msm.929347] [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: 12/20/2022] Open
Abstract
Background The aims of this study included 3 aspects: 1) assessing the expression of Apolipoprotein C1 (APOC1) in clear cell renal cell carcinoma (ccRCC) and normal groups; 2) evaluating the prognostic significance of APOC1 expression in the overall survival (OS) of ccRCC patients; and 3) exploring APOC1-related signaling pathways. Material/Methods The APOC1 expression value and clinical data of ccRCC patients were obtained from the cBioPortal database. We then evaluated the association of APOC1 expression with clinical characteristics of ccRCC patients. We also assessed the correlation between APOC1 expression and clinical outcome using Kaplan-Meier method. Our work then verified the independent prognostic factors of ccRCC by Cox regression analysis. Finally, the potential role of genes co-expressed with APOC1 was revealed via functional enrichment analysis. Results Bioinformatic data revealed that APOC1 was expressed at higher levels in ccRCC tissue than in the normal group (all P<0.05). The high expression of APOC1 was associated with unfavorable prognosis of female patients (P<0.01), but not of male patients. APOC1 high expression also shortened the survival time of ccRCC patients age ≥60 years old (P<0.05). Cox regression analysis further indicated that APOC1 expression was an independent prognostic factor for OS of ccRCC patients. Additionally, we found that APOC1 expression was significantly associated with sex, grade, clinical stage, and T stage. Finally, enrichment analysis suggested that APOC1-associated pathways were involved in tumor growth and metastasis. Conclusions The current study indicated that APOC1 was highly expressed in ccRCC and was significantly associated with key clinical features. APOC1 appears to be an independent prognostic factor in patients with ccRCC. Importantly, APOC1 might be a potential therapeutic target for ccRCC via regulating pathways involved in cell growth and metastasis.
Collapse
Affiliation(s)
- Huaying Xiao
- Department of Nephrology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Yifang Xu
- Department of Nephrology, Dongyang People's Hospital, Dongyang, Zhejiang, China (mainland)
| |
Collapse
|