1
|
Taha M, AlDuwaisan A, Daneshmand M, Ibrahim MM, Bourget-Murray J, Grammatopoulos G, Garceau S, Abdelbary H. Mapping Staphylococcus aureus at Early and Late Stages of Infection in a Clinically Representative Hip Prosthetic Joint Infection Rat Model. Microorganisms 2024; 12:1895. [PMID: 39338569 PMCID: PMC11433939 DOI: 10.3390/microorganisms12091895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Prosthetic joint infection (PJI) continues to be a devastating complication following total joint replacement surgeries where Staphylococcus aureus is the main offending organism. To improve our understanding of the disease pathogenesis, a histological analysis of infected peri-implant tissue in a hip PJI rat model was utilized to assess S. aureus spread and tissue reaction at early and late stages of infection. Sprague-Dawley rats were used and received a left cemented hip hemiarthroplasty using a 3D-printed titanium femoral stem. The rats received an intra-articular injection of S. aureus Xen36. These infected rats were sacrificed either at 3 days post-infection (early-stage infection) or at 13-days post-infection (late-stage infection). The femoral and acetabular tissues of all animals were harvested at euthanasia. Histological analysis for the harvested tissue was performed using immunohistochemistry, hematoxylin and eosin, as well as Masson's trichrome stains. Histological examination revealed significant quantitative and qualitative differences in peri-implant tissue response to infection at early and late stages. This hip PJI rat model identified clear histologic differences between early and late stages of S. aureus infection and how quickly bacterial infiltration could occur. These findings can provide insight into why certain surgical strategies like debridement and antibiotics may be associated with high failure rates.
Collapse
Affiliation(s)
- Mariam Taha
- Chronic Disease Program, The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Division of Orthopaedic Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Abdullah AlDuwaisan
- Division of Orthopaedic Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
- Division of Orthopaedic Surgery, Faculty of Medicine, Kuwait University, Jabriya, Kuwait
| | - Manijeh Daneshmand
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mazen M Ibrahim
- Division of Orthopaedic Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | | | | | - Simon Garceau
- Division of Orthopaedic Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Hesham Abdelbary
- Chronic Disease Program, The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Division of Orthopaedic Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
2
|
Slobodianyk-Kolomoiets M, Khlebas S, Mazur I, Rudnieva K, Potochilova V, Iungin O, Kamyshnyi O, Kamyshna I, Potters G, Spiers AJ, Moshynets O. Extracellular host DNA contributes to pathogenic biofilm formation during periodontitis. Front Cell Infect Microbiol 2024; 14:1374817. [PMID: 38779563 PMCID: PMC11109387 DOI: 10.3389/fcimb.2024.1374817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/09/2024] [Indexed: 05/25/2024] Open
Abstract
Introduction Periodontal diseases are known to be associated with polymicrobial biofilms and inflammasome activation. A deeper understanding of the subgingival cytological (micro) landscape, the role of extracellular DNA (eDNA) during periodontitis, and contribution of the host immune eDNA to inflammasome persistence, may improve our understanding of the mechanisms underlaying severe forms of periodontitis. Methods In this work, subgingival biolfilms developing on biologically neutral polyethylene terephthalate films placed in gingival cavities of patients with chronic periodontitis were investigated by confocal laser scanning microscopy (CLSM). This allowed examination of realistic cytological landscapes and visualization of extracellular polymeric substances (EPS) including amyloids, total proteins, carbohydrates and eDNA, as well as comparison with several single-strain in vitro model biofilms produced by oral pathogens such as Klebsiella pneumoniae, Pseudomonas aeruginosa, Staphylococcus aureus, Streptococcus gordonii, S. sanguinis and S. mitis. Fluorescence in situ hybridization (FISH) analysis was also used to identify eDNA derived from eubacteria, streptococci and members of the Bacteroides-Porphyromonas-Prevotella (BPP) group associated with periodontitis. Results Analysis of subgingival biofilm EPS revealed low levels of amyloids and high levels of eDNA which appears to be the main matrix component. However, bacterial eDNA contributed less than a third of the total eDNA observed, suggesting that host-derived eDNA released in neutrophil extracellular traps may be of more importance in the development of biofilms causing periodontitis. Discussion eDNA derived from host immunocompetent cells activated at the onset of periodontitis may therefore be a major driver of bacterial persistence and pathogenesis.
Collapse
Affiliation(s)
| | - Svitlana Khlebas
- Department of Dentistry, Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
| | - Iryna Mazur
- Department of Dentistry, Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
| | - Kateryna Rudnieva
- Central Clinical Diagnostic Laboratory, Kyiv Regional Clinical Hospital, Kyiv, Ukraine
- Department of Microbiology, Virology and Immunology, Bogomolets National Medical Academy, Kyiv, Ukraine
| | | | - Olga Iungin
- Biofilm Study Group, Department of Cell Regulatory Mechanisms, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
- Department of Biotechnology, Leather and Fur, Faculty of Chemical and Biopharmaceutical Technologies, Kyiv National University of Technologies and Design, Kyiv, Ukraine
| | - Olexandr Kamyshnyi
- Microbiology, Virology and Immunology Department, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Kamyshna
- Microbiology, Virology and Immunology Department, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Geert Potters
- Antwerp Maritime Academy, Antwerp, Belgium
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Andrew J. Spiers
- School of Applied Sciences, Abertay University, Dundee, United Kingdom
| | - Olena Moshynets
- Biofilm Study Group, Department of Cell Regulatory Mechanisms, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
3
|
Liu CG, Li DY, Gao X, Ma T, Zhang K, Liu DY. Examining the causal relationship between circulating immune cells and the susceptibility to osteomyelitis: A Mendelian randomization study. Int Immunopharmacol 2024; 131:111815. [PMID: 38492335 DOI: 10.1016/j.intimp.2024.111815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Osteomyelitis is considered as a deleterious inflammatory condition affecting the bone, primarily attributed to pathogenic infection. However, the underlying factors predisposing individuals to osteomyelitis remain incompletely elucidated. The immune system plays a multifaceted role in the progression of this condition, yet previous observational studies and randomized controlled trials investigating the association between circulating immune cell counts and osteomyelitis have been constrained. In order to address this knowledge gap, we conducted a Mendelian randomization (MR) analysis to evaluate the impact of diverse immune cell counts on the risk of developing osteomyelitis. METHODS In our study, we utilized single nucleotide polymorphisms (SNPs) that have been strongly linked to circulating immune cells or specific lymphocyte subtypes, as identified in large-scale genome-wide association studies (GWAS). These SNPs served as instrumental variables (IVs) for our MR analysis. We employed a more relaxed clumping threshold to conduct MR analysis on several related lymphocyte subtypes. To estimate causal effects, we utilized the Wald ratio, as well as the random-effects inverse variance weighted (IVW) and weighted median (WM) methods. To enhance the credibility of our results, we performed F-statistic calculations and a series of sensitivity analyses. RESULTS Our findings revealed a significant correlation between the absolute count of circulating lymphocytes and the risk of osteomyelitis [odds ratio(OR) 1.20;95 % confidence interval (CI), 1.08-1.32;P = 0.0005]. Furthermore, we identified a causal relationship between the absolute count of CD8+ T cells and susceptibility to osteomyelitis (OR 1.16; 95 % CI, 1.04-1.30; P = 0.0098). Importantly, these findings remained robust across a wide range of sensitivity analyses. CONCLUSION Through our MR analysis, we have provided evidence supporting a causal relationship between genetic predisposition to higher circulating immune cell counts and an increased risk of osteomyelitis. Specifically, our findings highlight the association between elevated CD8+ T cell counts and a heightened susceptibility to osteomyelitis. These results offer valuable insights for the future exploration of immunotherapy approaches in the management of osteomyelitis.
Collapse
Affiliation(s)
- Chun-Gui Liu
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Dong-Yang Li
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xi Gao
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Teng Ma
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Kun Zhang
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - De-Yin Liu
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
4
|
Konwar B, De S, Das G, Ramesh A. Inhibition of staphylococcal nuclease by benzimidazole-based Ligand: Implications in DNA-Mediated entrapment and uptake of MRSA by Macrophage-like cells. Bioorg Chem 2024; 144:107133. [PMID: 38278047 DOI: 10.1016/j.bioorg.2024.107133] [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: 10/04/2023] [Revised: 12/22/2023] [Accepted: 01/11/2024] [Indexed: 01/28/2024]
Abstract
The staphylococcal nuclease also referred as micrococcal nuclease (MNase) is a key drug target as the enzyme degrades the neutrophil extracellular trap (NET) and empowers the pathogen to subvert the host innate immune system. To this end, the current study presents a critical evaluation of MNase inhibition rendered by benzimidazole-based ligands (C1 and C2) and probes its therapeutic implications. A nuclease assay indicated that MNase inhibition rendered by C1 and C2 was ∼ 55 % and ∼ 72 %, respectively, at the highest tested concentration of 10 µM. Studies on enzyme kinetics revealed that C2 rendered non-competitive inhibition and significantly reduced MNase turnover number (Kcat) and catalytic efficiency (Kcat/Km) with an IC50 value of ∼ 1122 nM. In CD spectroscopy, a notable perturbation in the β-sheet content of MNase was observed in presence of C2. Fluorescence-microscope analysis indicated that MNase inhibition by C2 could restore entrapment of methicillin-resistant Staphylococcus aureus (MRSA) in calf-thymus DNA (CT-DNA). Flow cytometry and confocal microscope analysis revealed that uptake of DNA-entrapped MRSA by activated THP-1 cells was reinstated by MNase inhibition rendered by C2. Inhibition of nuclease by the non-toxic ligand C2 holds therapeutic prospect as it has the potential to bolster the DNA-mediated entrapment machinery and mitigate MRSA infections.
Collapse
Affiliation(s)
- Barlina Konwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Sagnik De
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Gopal Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Aiyagari Ramesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
5
|
Piuzzi NS, Klika AK, Lu Q, Higuera-Rueda CA, Stappenbeck T, Visperas A. Periprosthetic joint infection and immunity: Current understanding of host-microbe interplay. J Orthop Res 2024; 42:7-20. [PMID: 37874328 DOI: 10.1002/jor.25723] [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: 05/31/2023] [Revised: 09/19/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Periprosthetic joint infection (PJI) is a major complication of total joint arthroplasty. Even with current treatments, failure rates are unacceptably high with a 5-year mortality rate of 26%. Majority of the literature in the field has focused on development of better biomarkers for diagnostics and treatment strategies including innovate antibiotic delivery systems, antibiofilm agents, and bacteriophages. Nevertheless, the role of the immune system, our first line of defense during PJI, is not well understood. Evidence of infection in PJI patients is found within circulation, synovial fluid, and tissue and include numerous cytokines, metabolites, antimicrobial peptides, and soluble receptors that are part of the PJI diagnosis workup. Macrophages, neutrophils, and myeloid-derived suppressor cells (MDSCs) are initially recruited into the joint by chemokines and cytokines produced by immune cells and bacteria and are activated by pathogen-associated molecular patterns. While these cells are efficient killers of planktonic bacteria by phagocytosis, opsonization, degranulation, and recruitment of adaptive immune cells, biofilm-associated bacteria are troublesome. Biofilm is not only a physical barrier for the immune system but also elicits effector functions. Additionally, bacteria have developed mechanisms to evade the immune system by inactivating effector molecules, promoting killing or anti-inflammatory effector cell phenotypes, and intracellular persistence and dissemination. Understanding these shortcomings and the mechanisms by which bacteria can subvert the immune system may open new approaches to better prepare our own immune system to combat PJI. Furthermore, preoperative immune system assessment and screening for dysregulation may aid in developing preventative interventions to decrease PJI incidence.
Collapse
Affiliation(s)
- Nicolas S Piuzzi
- Department of Orthopaedic Surgery, Cleveland Clinic Adult Reconstruction Research (CCARR), Cleveland Clinic, Cleveland, Ohio, USA
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alison K Klika
- Department of Orthopaedic Surgery, Cleveland Clinic Adult Reconstruction Research (CCARR), Cleveland Clinic, Cleveland, Ohio, USA
| | - Qiuhe Lu
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Anabelle Visperas
- Department of Orthopaedic Surgery, Cleveland Clinic Adult Reconstruction Research (CCARR), Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
6
|
Arciola CR, Ravaioli S, Mirzaei R, Dolzani P, Montanaro L, Daglia M, Campoccia D. Biofilms in Periprosthetic Orthopedic Infections Seen through the Eyes of Neutrophils: How Can We Help Neutrophils? Int J Mol Sci 2023; 24:16669. [PMID: 38068991 PMCID: PMC10706149 DOI: 10.3390/ijms242316669] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Despite advancements in our knowledge of neutrophil responses to planktonic bacteria during acute inflammation, much remains to be elucidated on how neutrophils deal with bacterial biofilms in implant infections. Further complexity transpires from the emerging findings on the role that biomaterials play in conditioning bacterial adhesion, the variety of biofilm matrices, and the insidious measures that biofilm bacteria devise against neutrophils. Thus, grasping the entirety of neutrophil-biofilm interactions occurring in periprosthetic tissues is a difficult goal. The bactericidal weapons of neutrophils consist of the following: ready-to-use antibacterial proteins and enzymes stored in granules; NADPH oxidase-derived reactive oxygen species (ROS); and net-like structures of DNA, histones, and granule proteins, which neutrophils extrude to extracellularly trap pathogens (the so-called NETs: an allusive acronym for "neutrophil extracellular traps"). Neutrophils are bactericidal (and therefore defensive) cells endowed with a rich offensive armamentarium through which, if frustrated in their attempts to engulf and phagocytose biofilms, they can trigger the destruction of periprosthetic bone. This study speculates on how neutrophils interact with biofilms in the dramatic scenario of implant infections, also considering the implications of this interaction in view of the design of new therapeutic strategies and functionalized biomaterials, to help neutrophils in their arduous task of managing biofilms.
Collapse
Affiliation(s)
- Carla Renata Arciola
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Stefano Ravaioli
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Paolo Dolzani
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
| | - Lucio Montanaro
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy;
| | - Davide Campoccia
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| |
Collapse
|
7
|
Chen Y, Liu Z, Lin Z, Lu M, Fu Y, Liu G, Yu B. The effect of Staphylococcus aureus on innate and adaptive immunity and potential immunotherapy for S. aureus-induced osteomyelitis. Front Immunol 2023; 14:1219895. [PMID: 37744377 PMCID: PMC10517662 DOI: 10.3389/fimmu.2023.1219895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Osteomyelitis is a chronic inflammatory bone disease caused by infection of open fractures or post-operative implants. Particularly in patients with open fractures, the risk of osteomyelitis is greatly increased as the soft tissue damage and bacterial infection are often more severe. Staphylococcus aureus, one of the most common pathogens of osteomyelitis, disrupts the immune response through multiple mechanisms, such as biofilm formation, virulence factor secretion, and metabolic pattern alteration, which attenuates the effectiveness of antibiotics and surgical debridement toward osteomyelitis. In osteomyelitis, immune cells such as neutrophils, macrophages and T cells are activated in response to pathogenic bacteria invasion with excessive inflammatory factor secretion, immune checkpoint overexpression, and downregulation of immune pathway transcription factors, which enhances osteoclastogenesis and results in bone destruction. Therefore, the study of the mechanisms of abnormal immunity will be a new breakthrough in the treatment of osteomyelitis.
Collapse
Affiliation(s)
- Yingqi Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Zixian Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Zexin Lin
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Mincheng Lu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Yong Fu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
- Trauma Center, Department of Orthopaedic Trauma, The Second Affiliated Hospital of Hengyang Medical College, South China University, Hengyang, China
| | - Guanqiao Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| |
Collapse
|
8
|
Debourgogne A, Monpierre L, Sy KA, Valsecchi I, Decousser JW, Botterel F. Interactions between Bacteria and Aspergillus fumigatus in Airways: From the Mycobiome to Molecular Interactions. J Fungi (Basel) 2023; 9:900. [PMID: 37755008 PMCID: PMC10533028 DOI: 10.3390/jof9090900] [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: 07/31/2023] [Revised: 08/26/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023] Open
Abstract
Interactions between different kingdoms of microorganisms in humans are common but not well described. A recent analysis of the mycobiome has described the presence of different fungi and their positive and/or negative interactions with bacteria and other fungi. In chronic respiratory diseases, these different microorganisms form mixed biofilms to live inside. The interactions between Gram-negative bacteria and filamentous fungi in these biofilms have attracted more attention recently. In this review, we analyse the microbiota of the respiratory tract of healthy individuals and patients with chronic respiratory disease. Additionally, we describe the regulatory mechanisms that rule the mixed biofilms of Aspergillus fumigatus and Gram-negative bacteria and the effects of this biofilm on clinical presentations.
Collapse
Affiliation(s)
- Anne Debourgogne
- UR 7300, Stress Immunité Pathogène, Université de Lorraine, 54000 Vandoeuvre les Nancy, France;
| | - Lorra Monpierre
- Unité de Parasitologie-Mycologie, Département de Prévention, Diagnostic et Traitement des Infections, CHU Henri Mondor, Assistance Publique des Hôpitaux de Paris (APHP), 94000 Créteil, France;
- UR DYNAMYC 7380, Faculté de Santé, Univ Paris-Est Créteil (UPEC), Ecole Nationale Vétérinaire d’Alfort (ENVA), USC Anses, 94700 Créteil, France; (K.A.S.); (I.V.); (J.-W.D.)
| | - Khadeeja Adam Sy
- UR DYNAMYC 7380, Faculté de Santé, Univ Paris-Est Créteil (UPEC), Ecole Nationale Vétérinaire d’Alfort (ENVA), USC Anses, 94700 Créteil, France; (K.A.S.); (I.V.); (J.-W.D.)
- Institut National de la Santé et de la Recherche Médicale (Inserm) U955, 94010 Créteil, France
| | - Isabel Valsecchi
- UR DYNAMYC 7380, Faculté de Santé, Univ Paris-Est Créteil (UPEC), Ecole Nationale Vétérinaire d’Alfort (ENVA), USC Anses, 94700 Créteil, France; (K.A.S.); (I.V.); (J.-W.D.)
| | - Jean-Winoc Decousser
- UR DYNAMYC 7380, Faculté de Santé, Univ Paris-Est Créteil (UPEC), Ecole Nationale Vétérinaire d’Alfort (ENVA), USC Anses, 94700 Créteil, France; (K.A.S.); (I.V.); (J.-W.D.)
- Department of Infection Control, University Hospital Henri Mondor, Assistance Publique—Hôpitaux de Paris, 94000 Créteil, France
| | - Françoise Botterel
- Unité de Parasitologie-Mycologie, Département de Prévention, Diagnostic et Traitement des Infections, CHU Henri Mondor, Assistance Publique des Hôpitaux de Paris (APHP), 94000 Créteil, France;
- UR DYNAMYC 7380, Faculté de Santé, Univ Paris-Est Créteil (UPEC), Ecole Nationale Vétérinaire d’Alfort (ENVA), USC Anses, 94700 Créteil, France; (K.A.S.); (I.V.); (J.-W.D.)
| |
Collapse
|
9
|
Perry EK, Tan MW. Bacterial biofilms in the human body: prevalence and impacts on health and disease. Front Cell Infect Microbiol 2023; 13:1237164. [PMID: 37712058 PMCID: PMC10499362 DOI: 10.3389/fcimb.2023.1237164] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/11/2023] [Indexed: 09/16/2023] Open
Abstract
Bacterial biofilms can be found in most environments on our planet, and the human body is no exception. Consisting of microbial cells encased in a matrix of extracellular polymers, biofilms enable bacteria to sequester themselves in favorable niches, while also increasing their ability to resist numerous stresses and survive under hostile circumstances. In recent decades, biofilms have increasingly been recognized as a major contributor to the pathogenesis of chronic infections. However, biofilms also occur in or on certain tissues in healthy individuals, and their constituent species are not restricted to canonical pathogens. In this review, we discuss the evidence for where, when, and what types of biofilms occur in the human body, as well as the diverse ways in which they can impact host health under homeostatic and dysbiotic states.
Collapse
Affiliation(s)
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech, South San Francisco, CA, United States
| |
Collapse
|
10
|
Taha M, Arnaud T, Lightly TJ, Peters D, Wang L, Chen W, Cook BWM, Theriault SS, Abdelbary H. Combining bacteriophage and vancomycin is efficacious against MRSA biofilm-like aggregates formed in synovial fluid. Front Med (Lausanne) 2023; 10:1134912. [PMID: 37359001 PMCID: PMC10289194 DOI: 10.3389/fmed.2023.1134912] [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: 12/31/2022] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Background Biofilm formation is a major clinical challenge contributing to treatment failure of periprosthetic joint infection (PJI). Lytic bacteriophages (phages) can target biofilm associated bacteria at localized sites of infection. The aim of this study is to investigate whether combination therapy of phage and vancomycin is capable of clearing Staphylococcus aureus biofilm-like aggregates formed in human synovial fluid. Methods In this study, S. aureus BP043, a PJI clinical isolate was utilized. This strain is a methicillin-resistant S. aureus (MRSA) biofilm-former. Phage Remus, known to infect S. aureus, was selected for the treatment protocol. BP043 was grown as aggregates in human synovial fluid. The characterization of S. aureus aggregates was assessed for structure and size using scanning electron microscopy (SEM) and flow cytometry, respectively. Moreover, the formed aggregates were subsequently treated in vitro with: (a) phage Remus [∼108 plaque-forming units (PFU)/ml], (b) vancomycin (500 μg/ml), or (c) phage Remus (∼108 PFU/ml) followed by vancomycin (500 μg/ml), for 48 h. Bacterial survival was quantified by enumeration [colony-forming units (CFU)/ml]. The efficacy of phage and vancomycin against BP043 aggregates was assessed in vivo as individual treatments and in combination. The in vivo model utilized Galleria mellonella larvae which were infected with BP043 aggregates pre-formed in synovial fluid. Results Scanning electron microscopy (SEM) images and flow cytometry data demonstrated the ability of human synovial fluid to promote formation of S. aureus aggregates. Treatment with Remus resulted in significant reduction in viable S. aureus residing within the synovial fluid aggregates compared to the aggregates that did not receive Remus (p < 0.0001). Remus was more efficient in eliminating viable bacteria within the aggregates compared to vancomycin (p < 0.0001). Combination treatment of Remus followed by vancomycin was more efficacious in reducing bacterial load compared to using either Remus or vancomycin alone (p = 0.0023, p < 0.0001, respectively). When tested in vivo, this combination treatment also resulted in the highest survival rate (37%) 96 h post-treatment, compared to untreated larvae (3%; p < 0.0001). Conclusion We demonstrate that combining phage Remus and vancomycin led to synergistic interaction against MRSA biofilm-like aggregates in vitro and in vivo.
Collapse
Affiliation(s)
- Mariam Taha
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Orthopedic Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Tia Arnaud
- Cytophage Technologies Inc., Winnipeg, MB, Canada
- Department of Microbiology, The University of Manitoba, Winnipeg, MB, Canada
| | | | - Danielle Peters
- Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON, Canada
| | - Liyuan Wang
- Cell Biology and Image Acquisition, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON, Canada
- Department of Biology, Brock University, St. Catharines, ON, Canada
| | | | - Steven S. Theriault
- Cytophage Technologies Inc., Winnipeg, MB, Canada
- Department of Microbiology, The University of Manitoba, Winnipeg, MB, Canada
| | - Hesham Abdelbary
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Orthopedic Surgery, The Ottawa Hospital, Ottawa, ON, Canada
- Department of Surgery, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
11
|
Maimaiti Z, Li Z, Xu C, Fu J, Hao LB, Chen JY, Chai W. Host Immune Regulation in Implant-Associated Infection (IAI): What Does the Current Evidence Provide Us to Prevent or Treat IAI? Bioengineering (Basel) 2023; 10:bioengineering10030356. [PMID: 36978747 PMCID: PMC10044746 DOI: 10.3390/bioengineering10030356] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
The number of orthopedic implants for bone fixation and joint arthroplasty has been steadily increasing over the past few years. However, implant-associated infection (IAI), a major complication in orthopedic surgery, impacts the quality of life and causes a substantial economic burden on patients and societies. While research and study on IAI have received increasing attention in recent years, the failure rate of IAI has still not decreased significantly. This is related to microbial biofilms and their inherent antibiotic resistance, as well as the various mechanisms by which bacteria evade host immunity, resulting in difficulties in diagnosing and treating IAIs. Hence, a better understanding of the complex interactions between biofilms, implants, and host immunity is necessary to develop new strategies for preventing and controlling these infections. This review first discusses the challenges in diagnosing and treating IAI, followed by an extensive review of the direct effects of orthopedic implants, host immune function, pathogenic bacteria, and biofilms. Finally, several promising preventive or therapeutic alternatives are presented, with the hope of mitigating or eliminating the threat of antibiotic resistance and refractory biofilms in IAI.
Collapse
Affiliation(s)
- Zulipikaer Maimaiti
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhuo Li
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Chi Xu
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jun Fu
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Li-Bo Hao
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Ji-Ying Chen
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
- Correspondence: (J.-Y.C.); (W.C.)
| | - Wei Chai
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
- Correspondence: (J.-Y.C.); (W.C.)
| |
Collapse
|
12
|
Aristotelous AC. Biofilm neutrophils interactions under hypoxia: A mathematical modeling study. Math Biosci 2022; 352:108893. [PMID: 36029807 DOI: 10.1016/j.mbs.2022.108893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/09/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022]
Abstract
Neutrophils are important to the defense of the host against bacterial infection. Pathogens and the immune system cells create via respiration, a hypoxic environment in infected regions. Hypoxic conditions affect both the neutrophil's ability to eradicate the infection and also change the behavior of the bacterial-pathogens by eliciting the production of various virulence factors, the creation of bacterial biofilm and the initialization of anaerobic metabolism. In this work interactions of bacterial biofilm and neutrophils are studied in a domain where oxygen is diffusing into the environment and is being consumed by biofilm. Within a hypoxic environment, bacteria grow anaerobically and secrete higher levels of toxin that diffuses and lyses neutrophils. A mathematical model explicitly representing the biofilm volume fraction, oxygen, and diffusive virulence factors (toxin) as well as killing of bacteria by neutrophils is developed and studied first in 1D and then in 2D. Stability analysis and numerical simulations showing the effects of oxygen and toxin concentration on neutrophil-bacteria interactions are presented to identify different possible scenarios that can lead to elimination of the infection or its persistence as a chronic infection. Specifically, when bacteria are allowed to utilize anaerobic breathing and or to produce toxin, their fitness is enhanced against neutrophils attacks. A possible insight on how virulent bacterial colonies can synergistically resist neutrophils and survive is presented.
Collapse
Affiliation(s)
- Andreas C Aristotelous
- Department of Mathematics, Buchtel College of Arts and Sciences, The University of Akron, Akron, OH, 44325-4002, USA.
| |
Collapse
|
13
|
Meroni G, Tsikopoulos A, Tsikopoulos K, Allemanno F, Martino PA, Soares Filipe JF. A Journey into Animal Models of Human Osteomyelitis: A Review. Microorganisms 2022; 10:1135. [PMID: 35744653 PMCID: PMC9228829 DOI: 10.3390/microorganisms10061135] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 12/24/2022] Open
Abstract
Osteomyelitis is an infection of the bone characterized by progressive inflammatory destruction and apposition of new bone that can spread via the hematogenous route (hematogenous osteomyelitis (HO)), contiguous spread (contiguous osteomyelitis (CO)), and direct inoculation (osteomyelitis associated with peripheral vascular insufficiency (PVI)). Given the significant financial burden posed by osteomyelitis patient management, the development of new preventive and treatment methods is warranted. To achieve this objective, implementing animal models (AMs) of infection such as rats, mice, rabbits, avians, dogs, sheep, goats, and pigs might be of the essence. This review provides a literature analysis of the AMs developed and used to study osteomyelitis. Historical relevance and clinical applicability were taken into account to choose the best AMs, and some study methods are briefly described. Furthermore, the most significant strengths and limitations of each species as AM are discussed, as no single model incorporates all features of osteomyelitis. HO's clinical manifestation results in extreme variability between patients due to multiple variables (e.g., age, sex, route of infection, anatomical location, and concomitant diseases) that could alter clinical studies. However, these variables can be controlled and tested through different animal models.
Collapse
Affiliation(s)
- Gabriele Meroni
- One Health Unit, Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy; (F.A.); (P.A.M.)
| | - Alexios Tsikopoulos
- Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | | | - Francesca Allemanno
- One Health Unit, Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy; (F.A.); (P.A.M.)
| | - Piera Anna Martino
- One Health Unit, Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy; (F.A.); (P.A.M.)
| | - Joel Fernando Soares Filipe
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy;
| |
Collapse
|
14
|
Caldara M, Belgiovine C, Secchi E, Rusconi R. Environmental, Microbiological, and Immunological Features of Bacterial Biofilms Associated with Implanted Medical Devices. Clin Microbiol Rev 2022; 35:e0022120. [PMID: 35044203 PMCID: PMC8768833 DOI: 10.1128/cmr.00221-20] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The spread of biofilms on medical implants represents one of the principal triggers of persistent and chronic infections in clinical settings, and it has been the subject of many studies in the past few years, with most of them focused on prosthetic joint infections. We review here recent works on biofilm formation and microbial colonization on a large variety of indwelling devices, ranging from heart valves and pacemakers to urological and breast implants and from biliary stents and endoscopic tubes to contact lenses and neurosurgical implants. We focus on bacterial abundance and distribution across different devices and body sites and on the role of environmental features, such as the presence of fluid flow and properties of the implant surface, as well as on the interplay between bacterial colonization and the response of the human immune system.
Collapse
Affiliation(s)
- Marina Caldara
- Interdepartmental Center on Safety, Technologies, and Agri-food Innovation (SITEIA.PARMA), University of Parma, Parma, Italy
| | - Cristina Belgiovine
- IRCCS Humanitas Research Hospital, Rozzano–Milan, Italy
- Scuola di Specializzazione in Microbiologia e Virologia, Università degli Studi di Pavia, Pavia, Italy
| | - Eleonora Secchi
- Institute of Environmental Engineering, ETH Zürich, Zürich, Switzerland
| | - Roberto Rusconi
- IRCCS Humanitas Research Hospital, Rozzano–Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele–Milan, Italy
| |
Collapse
|
15
|
The Influence of Antibiotic Resistance on Innate Immune Responses to Staphylococcus aureus Infection. Antibiotics (Basel) 2022; 11:antibiotics11050542. [PMID: 35625186 PMCID: PMC9138074 DOI: 10.3390/antibiotics11050542] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 11/16/2022] Open
Abstract
Staphylococcus aureus (S. aureus) causes a broad range of infections and is associated with significant morbidity and mortality. S. aureus produces a diverse range of cellular and extracellular factors responsible for its invasiveness and ability to resist immune attack. In recent years, increasing resistance to last-line anti-staphylococcal antibiotics daptomycin and vancomycin has been observed. Resistant strains of S. aureus are highly efficient in invading a variety of professional and nonprofessional phagocytes and are able to survive inside host cells. Eliciting immune protection against antibiotic-resistant S. aureus infection is a global challenge, requiring both innate and adaptive immune effector mechanisms. Dendritic cells (DC), which sit at the interface between innate and adaptive immune responses, are central to the induction of immune protection against S. aureus. However, it has been observed that S. aureus has the capacity to develop further antibiotic resistance and acquire increased resistance to immunological recognition by the innate immune system. In this article, we review the strategies utilised by S. aureus to circumvent antibiotic and innate immune responses, especially the interaction between S. aureus and DC, focusing on how this relationship is perturbed with the development of antibiotic resistance.
Collapse
|
16
|
Forson AM, Rosman CWK, van Kooten TG, van der Mei HC, Sjollema J. Micrococcal Nuclease stimulates Staphylococcus aureus Biofilm Formation in a Murine Implant Infection Model. Front Cell Infect Microbiol 2022; 11:799845. [PMID: 35111695 PMCID: PMC8801922 DOI: 10.3389/fcimb.2021.799845] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/21/2021] [Indexed: 12/22/2022] Open
Abstract
Advancements in contemporary medicine have led to an increasing life expectancy which has broadened the application of biomaterial implants. As each implant procedure has an innate risk of infection, the number of biomaterial-associated infections keeps rising. Staphylococcus aureus causes 34% of such infections and is known as a potent biofilm producer. By secreting micrococcal nuclease S. aureus is able to escape neutrophil extracellular traps by cleaving their DNA-backbone. Also, micrococcal nuclease potentially limits biofilm growth and adhesion by cleaving extracellular DNA, an important constituent of biofilms. This study aimed to evaluate the impact of micrococcal nuclease on infection persistence and biofilm formation in a murine biomaterial-associated infection-model with polyvinylidene-fluoride mesh implants inoculated with bioluminescent S. aureus or its isogenic micrococcal nuclease deficient mutant. Supported by results based on in-vivo bioluminescence imaging, ex-vivo colony forming unit counts, and histological analysis it was found that production of micrococcal nuclease enables S. aureus bacteria to evade the immune response around an implant resulting in a persistent infection. As a novel finding, histological analysis provided clear indications that the production of micrococcal nuclease stimulates S. aureus to form biofilms, the presence of which extended neutrophil extracellular trap formation up to 13 days after mesh implantation. Since micrococcal nuclease production appeared vital for the persistence of S. aureus biomaterial-associated infection, targeting its production could be a novel strategy in preventing biomaterial-associated infection.
Collapse
|
17
|
Rosman CWK, van Dijl JM, Sjollema J. Interactions between the foreign body reaction and Staphylococcus aureus biomaterial-associated infection. Winning strategies in the derby on biomaterial implant surfaces. Crit Rev Microbiol 2021; 48:624-640. [PMID: 34879216 DOI: 10.1080/1040841x.2021.2011132] [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: 02/06/2023]
Abstract
Biomaterial-associated infections (BAIs) are an increasing problem where antibiotic therapies are often ineffective. The design of novel strategies to prevent or combat infection requires a better understanding of how an implanted foreign body prevents the immune system from eradicating surface-colonizing pathogens. The objective of this review is to chart factors resulting in sub-optimal clearance of Staphylococcus aureus bacteria involved in BAIs. To this end, we first describe three categories of bacterial mechanisms to counter the host immune system around foreign bodies: direct interaction with host cells, modulation of intercellular communication, and evasion of the immune system. These mechanisms take place in a time frame that differentiates sterile foreign body reactions, BAIs, and soft tissue infections. In addition, we identify experimental interventions in S. aureus BAI that may impact infectious mechanisms. Most experimental treatments modulate the host response to infection or alter the course of BAI through implant surface modulation. In conclusion, the first week after implantation and infection is crucial for the establishment of an S. aureus biofilm that resists the local immune reaction and antibiotic treatment. Although established and chronic S. aureus BAI is still treatable and manageable, the focus of interventions should lie on this first period.
Collapse
Affiliation(s)
- Colin W K Rosman
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jelmer Sjollema
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
18
|
Hamilton J, Mohamed M, Witt B, Wimmer M, Shafikhani S. Therapeutic assessment of N-formyl-methionyl-leucyl-phenylalanine (fMLP) in reducing periprosthetic joint infection. Eur Cell Mater 2021; 42:122-138. [PMID: 34435345 PMCID: PMC8459619 DOI: 10.22203/ecm.v042a09] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Despite many preventive measures, including prophylactic antibiotics, periprosthetic joint infection (PJI) remains a devastating complication following arthroplasty, leading to pain, suffering, morbidity and substantial economic burden. Humans have a powerful innate immune system that can effectively control infections, if alerted quickly. Unfortunately, pathogens use many mechanisms to dampen innate immune responses. The study hypothesis was that immunomodulators that can jumpstart and direct innate immune responses (particularly neutrophils) at the surgical site of implant placement would boost immune responses and reduce PJI, even in the absence of antibiotics. To test this hypothesis, N-formyl-methionyl-leucyl-phenylalanine (fMLP) (a potent chemoattractant for phagocytic leukocytes including neutrophils) was used in a mouse model of PJI with Staphylococcus aureus (S. aureus). Mice receiving intramedullary femoral implants were divided into three groups: i) implant alone; ii) implant + S. aureus; iii) implant + fMLP + S. aureus. fMLP treatment reduced S. aureus infection levels by ~ 2-Log orders at day 3. Moreover, fMLP therapy reduced infection-induced peri-implant periosteal reaction, focal cortical loss and areas of inflammatory infiltrate in mice distal femora at day 10. Finally, fMLP treatment reduced pain behaviour and increased weight-bearing at the implant leg in infected mice at day 10. Data indicated that fMLP therapy is a promising novel approach for reducing PJI, if administered locally at surgical sites. Future work will be toward further enhancement and optimisation of an fMLP-based therapeutic approach through combination with antibiotics and/or implant coating with fMLP.
Collapse
Affiliation(s)
- J.L. Hamilton
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612-3806, USA,Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612-3806, USA
| | - M.F. Mohamed
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612-3806, USA
| | - B.R. Witt
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612-3806, USA
| | - M.A. Wimmer
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612-3806, USA,Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612-3806, USA
| | - S.H. Shafikhani
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612-3806, USA,Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612-3806, USA,Cancer Center, Rush University Medical Center, Chicago, IL 60612-3806, USA,Address for correspondence: Sasha H. Shafikhani, Department of Medicine, Division of Hematology, Oncology and Cell Therapy, Department of Microbial Pathogens and Immunity, Cancer Center, Rush University Medical Center, 1735 W. Harrison Street, Chicago, IL 60612-3806, USA. Telephone number: +1 3129421368
| |
Collapse
|
19
|
Pettygrove BA, Kratofil RM, Alhede M, Jensen PØ, Newton M, Qvortrup K, Pallister KB, Bjarnsholt T, Kubes P, Voyich JM, Stewart PS. Delayed neutrophil recruitment allows nascent Staphylococcus aureus biofilm formation and immune evasion. Biomaterials 2021; 275:120775. [PMID: 34243039 DOI: 10.1016/j.biomaterials.2021.120775] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/19/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023]
Abstract
Biofilms that form on implanted medical devices cause recalcitrant infections. The early events enabling contaminating bacteria to evade immune clearance, before a mature biofilm is established, are poorly understood. Live imaging in vitro demonstrated that Staphylococcus aureus sparsely inoculated on an abiotic surface can go undiscovered by human neutrophils, grow, and form aggregates. Small (~50 μm2) aggregates of attached bacteria resisted killing by human neutrophils, resulting in neutrophil lysis and bacterial persistence. In vivo, neutrophil recruitment to a peritoneal implant was spatially heterogenous, with some bacterial aggregates remaining undiscovered by neutrophils after 24 h. Intravital imaging in mouse skin revealed that attached S. aureus aggregates grew and remained undiscovered by neutrophils for up to 3 h. These results suggest a model in which delayed recruitment of neutrophils to an abiotic implant presents a critical window in which bacteria establish a nascent biofilm and acquire tolerance to neutrophil killing.
Collapse
Affiliation(s)
- Brian A Pettygrove
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA; Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Rachel M Kratofil
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Maria Alhede
- Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Peter Ø Jensen
- Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark; Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Michelle Newton
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Klaus Qvortrup
- Department of Biomedical Sciences/CFIM, University of Copenhagen, Copenhagen, Denmark
| | - Kyler B Pallister
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Paul Kubes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Jovanka M Voyich
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Philip S Stewart
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA; Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, USA.
| |
Collapse
|
20
|
Dauros-Singorenko P, Wiles S, Swift S. Staphylococcus aureus Biofilms and Their Response to a Relevant in vivo Iron Source. Front Microbiol 2020; 11:509525. [PMID: 33408695 PMCID: PMC7779473 DOI: 10.3389/fmicb.2020.509525] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 11/23/2020] [Indexed: 11/29/2022] Open
Abstract
Biofilm infections can be chronic, life threatening and challenging to eradicate. Understanding in vivo stimuli affecting the biofilm cycle is one step toward targeted prevention strategies. Iron restriction by the host is a stimulus for biofilm formation for some Staphylococcus aureus isolates; however, in some infection scenarios bacteria are exposed to abundant amounts of hemoglobin (Hb), which S. aureus is able to use as iron source. Thus, we hypothesized a role for Hb in the biofilm infection. Microplate “biofilm” assays showed biofilm-matrix production was increased in the presence of hemoglobin when compared to the provision of iron as an inorganic salt. Microscopic analysis of biofilms showed that the provision of iron as hemoglobin consistently caused thicker and more structured biofilms when compared to the effect of the inorganic iron source. Iron responsive biofilm gene expression analysis showed that Agr Quorum Sensing, a known biofilm dispersal marker, was repressed with hemoglobin but induced with an equivalent amount of inorganic iron in the laboratory strain Newman. The gene expression of two biofilm structuring agents, PSMα and PSMβ, differed in the response to the iron source provided and was not correlated to hemoglobin-structured biofilms. A comparison of the model pathogen S. aureus Newman with local clinical isolates demonstrated that while there was a similar phenotypic biofilm response to hemoglobin, there was substantial variation in the expression of key biofilm dispersal markers, suggesting an underappreciated variation in biofilm regulome among S. aureus isolates and that no general inferences can be made by studying the behavior of single strains.
Collapse
Affiliation(s)
- Priscila Dauros-Singorenko
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Siouxsie Wiles
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Simon Swift
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
21
|
Gries CM, Rivas Z, Chen J, Lo DD. Intravital Multiphoton Examination of Implant-Associated Staphylococcus aureus Biofilm Infection. Front Cell Infect Microbiol 2020; 10:574092. [PMID: 33178628 PMCID: PMC7593243 DOI: 10.3389/fcimb.2020.574092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
Bacterial infections associated with implanted medical devices represents a healthcare crisis due to their persistence, antibiotic tolerance, and immune avoidance. Indwelling devices are rapidly coated with host plasma and extracellular matrix proteins which can then be exploited by bacterial pathogens for adherence and subsequent biofilm development. Our understanding of the host-pathogen interface that determines the fate of biofilm-mediated infections is limited to the experimental models employed by laboratories studying these organisms. Current in vivo models of biofilm-mediated infection, while certainly useful, are typically limited to end-point analyses of bacterial burden enumeration, immune cell profiling, and cytokine/chemokine analysis. Thus, with these models, the complex, real-time assessment of biofilm development and innate immune cell activity remains imperceptible. Here, we describe a novel murine biofilm infection model employing time-lapse intravital multiphoton microscopy which permits concurrent and real-time visualization of Staphylococcus aureus biofilm formation and immune cell activity. Using cell tracking, we found that S. aureus biofilms impede neutrophil chemotaxis, redirecting their migration patterns to prevent biofilm invasion. This approach is the first to directly examine device-associated biofilm development and host-pathogen interactions and will serve to both further our understanding of infection development and help reveal the effects of future antibiofilm treatment strategies.
Collapse
Affiliation(s)
- Casey M Gries
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Zuivanna Rivas
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Justin Chen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - David D Lo
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
22
|
Kavanaugh JS, Leidal KG, Nauseef WM, Horswill AR. Cathepsin G Degrades Staphylococcus aureus Biofilms. J Infect Dis 2020; 223:1865-1869. [PMID: 32995850 DOI: 10.1093/infdis/jiaa612] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/25/2020] [Indexed: 12/22/2022] Open
Abstract
Polymorphonuclear leukocytes (PMN) phagocytose and kill individual bacteria but are far less efficient when challenged with bacterial aggregates. Consequently, growth within a biofilm affords Staphylococcus aureus some protection but PMN penetrate S. aureus biofilms and phagocytose bacteria, suggesting that enzymes released through neutrophil degranulation degrade biofilms into fragments small enough for phagocytosis. Here we show that the capacity of PMN to invade biofilms depended largely on the activity of secreted cathepsin G.
Collapse
Affiliation(s)
- Jeffrey S Kavanaugh
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kevin G Leidal
- Inflammation Program, Department of Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - William M Nauseef
- Inflammation Program, Department of Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Veterans Affairs, Eastern Colorado Health Care System, Denver, Colorado, USA
| |
Collapse
|
23
|
Maali Y, Journo C, Mahieux R, Dutartre H. Microbial Biofilms: Human T-cell Leukemia Virus Type 1 First in Line for Viral Biofilm but Far Behind Bacterial Biofilms. Front Microbiol 2020; 11:2041. [PMID: 33042035 PMCID: PMC7523422 DOI: 10.3389/fmicb.2020.02041] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/03/2020] [Indexed: 12/25/2022] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is a retrovirus associated with adult T-cell leukemia (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). To date, it is the unique published example of a virus able to form a biofilm at the surface of infected cells. Deeply studied in bacteria, bacterial biofilms represent multicellular assemblies of bacteria in contact with a surface and shielded by the extracellular matrix (ECM). Microbial lifestyle in biofilms, either viral or bacterial, is opposed structurally and physiologically to an isolated lifestyle, in which viruses or bacteria freely float in their environment. HTLV-1 biofilm formation is believed to be promoted by viral proteins, mainly Tax, through remodeling of the ECM of the infected cells. HTLV-1 biofilm has been linked to cell-to-cell transmission of the virus. However, in comparison to bacterial biofilms, very little is known on kinetics of viral biofilm formation or dissemination, but also on its pathophysiological roles, such as escape from immune detection or therapeutic strategies, as well as promotion of leukemogenesis. The switch between production of cell-free isolated virions and cell-associated viral biofilm, although not fully apprehended yet, remains a key step to understand HTLV-1 infection and pathogenesis.
Collapse
Affiliation(s)
- Yousef Maali
- CIRI - Centre International de Recherche en Infectiologie, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
| | - Chloé Journo
- CIRI - Centre International de Recherche en Infectiologie, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
| | - Renaud Mahieux
- CIRI - Centre International de Recherche en Infectiologie, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
| | - Hélène Dutartre
- CIRI - Centre International de Recherche en Infectiologie, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Lyon, France
| |
Collapse
|
24
|
Alhede M, Lorenz M, Fritz BG, Jensen PØ, Ring HC, Bay L, Bjarnsholt T. Bacterial aggregate size determines phagocytosis efficiency of polymorphonuclear leukocytes. Med Microbiol Immunol 2020; 209:669-680. [PMID: 32880037 PMCID: PMC7568703 DOI: 10.1007/s00430-020-00691-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
The ability of bacteria to aggregate and form biofilms impairs phagocytosis by polymorphonuclear leukocytes (PMNs). The aim of this study was to examine if the size of aggregates is critical for successful phagocytosis and how bacterial biofilms evade phagocytosis. We investigated the live interaction between PMNs and Pseudomonas aeruginosa, Staphylococcus aureus, Escherichia coli and Staphylococcus epidermidis using confocal scanning laser microscopy. Aggregate size significantly affected phagocytosis outcome and larger aggregates were less likely to be phagocytized. Aggregates of S. epidermidis were also less likely to be phagocytized than equally-sized aggregates of the other three species. We found that only aggregates of approx. 5 μm diameter or smaller were consistently phagocytosed. We demonstrate that planktonic and aggregated cells of all four species significantly reduced the viability of PMNs after 4 h of incubation. Our results indicate that larger bacterial aggregates are less likely to be phagocytosed by PMNs and we propose that, if the aggregates become too large, circulating PMNs may not be able to phagocytose them quickly enough, which may lead to chronic infection.
Collapse
Affiliation(s)
- Maria Alhede
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Melanie Lorenz
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Blaine Gabriel Fritz
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Peter Østrup Jensen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark.,Department of Clinical Microbiology, Rigshospitalet, Afsnit 9301, Juliane Maries Vej 22, DK-2100, Copenhagen Ø, Denmark.,Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, 2100, Copenhagen, Denmark
| | - Hans Christian Ring
- Department of Dermatology, Bispebjerg Hospital, Nielsine Nielsens Vej 9, København, NV, Denmark
| | - Lene Bay
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark. .,Department of Clinical Microbiology, Rigshospitalet, Afsnit 9301, Juliane Maries Vej 22, DK-2100, Copenhagen Ø, Denmark.
| |
Collapse
|
25
|
Shlepotina NM, Peshikova MV, Kolesnikov OL, Shishkova YS. Modern Conceptions about the Mechanisms of Interaction Between Biofilm and Cellular Immunity Factors. ACTA ACUST UNITED AC 2020. [DOI: 10.36233/0372-9311-2020-97-1-83-90] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Features of the cellular immune response in the presence of a microbial biofilm are well described in the literature. Based on numerous studies, it became possible to establish a number of patterns: mature biofilms are better protected from immune factors, the effectiveness of antibiofilm strategies depends on species of the microorganisms, forming the biofilm, and, accordingly, on the composition of the biopolymer matrix. For example, rhamnolipids and alginate of Pseudomonas aeruginosa exert a significant negative effect on the function of immunocompetent cells. The bacteria of biofilms became able to turn to their advantage many of the protective reactions developed by the immune system and fixed evolutionarily, applying them for the growth and development of the microbial consortium.
Collapse
|
26
|
Kim JE, Monmai C, Rod-in W, Jang AY, You SG, Lee SM, Jung SK, Park WJ. Co-immunomodulatory Activities of Anionic Macromolecules Extracted from Codium fragile with Red Ginseng Extract on Peritoneal Macrophage of Immune-Suppressed Mice. J Microbiol Biotechnol 2020; 30:352-358. [PMID: 31893613 PMCID: PMC9728336 DOI: 10.4014/jmb.1909.09062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this study we investigated the immune effects of oral administration of anionic macromolecules extracted from Codium fragile (CFAM) and red ginseng extract mixture on the peritoneal macrophage cells in immune-suppressed mice. Cyclophosphamide (CY) induces the immune-suppressed condition. CY-treated mice were orally fed with different concentrations of CFAM supplemented with red ginseng extract and the peritoneal macrophages collected. CY treatment significantly decreased the immune activities of peritoneal macrophages, compared to the normal mice. The administration of CFAM mixed with red ginseng extract significantly boosted the viability of macrophage cells and nitric oxide production of peritoneal macrophages. Further, the oral administration of CFAM mixed with red ginseng extract up-regulated the expression of iNOS, COX-2, and TLR-4 as well as cytokines such as IL-1β, IL-6, TNF-α, and IFN-γ more than the red ginseng-treated group. This study showed that CFAM enhanced the immune activity of red ginseng extract in the peritoneal macrophage cells of immune-suppressed mice. Furthermore, CFAM might be used as a co-stimulant of red ginseng extract through the regulation of macrophage cells for the enhancement of human health and immunity.
Collapse
Affiliation(s)
- Ji Eun Kim
- Department of Wellness-Bio Industry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
| | - Chaiwat Monmai
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung 5457, Republic of Korea
| | - Weerawan Rod-in
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung 5457, Republic of Korea
| | - A-yeong Jang
- Department of Wellness-Bio Industry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea,Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung 5457, Republic of Korea
| | - Sang-Guan You
- Department of Wellness-Bio Industry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea,Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung 5457, Republic of Korea
| | - Sang-min Lee
- Department of Wellness-Bio Industry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea,Department of Marine Biotechnology, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
| | - Seok-Kyu Jung
- Department of Horticulture, Daegu Catholic University, Gyeongsan 3830, Republic of Korea
| | - Woo Jung Park
- Department of Wellness-Bio Industry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea,Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung 5457, Republic of Korea,Corresponding author Phone: +82-33-640-2857 Fax: +82-33-640-2850 E-mail:
| |
Collapse
|
27
|
de Vor L, Rooijakkers SHM, van Strijp JAG. Staphylococci evade the innate immune response by disarming neutrophils and forming biofilms. FEBS Lett 2020; 594:2556-2569. [PMID: 32144756 DOI: 10.1002/1873-3468.13767] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/30/2020] [Accepted: 02/22/2020] [Indexed: 12/24/2022]
Abstract
Staphylococcus aureus and Staphylococcus epidermidis can cause many types of infections, ranging from skin infections to implant-associated infections. The primary innate immune response against bacterial infections involves complement activation, recruitment of phagocytes (most importantly neutrophils), and subsequent killing of the pathogen. However, staphylococci are not innocent bystanders; they actively obstruct this immune attack. To do that, S. aureus secretes several immune-evasion proteins to resist attack by the innate immune system. Furthermore, S. aureus and S. epidermidis are known for their ability to form biofilms on implanted medical devices and host tissues, which provides another important immune-evasion mechanism. Understanding these different strategies to resist immune attack will help to develop novel therapies against staphylococcal infections.
Collapse
Affiliation(s)
- Lisanne de Vor
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Suzan H M Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, The Netherlands
| |
Collapse
|
28
|
Hofstee MI, Muthukrishnan G, Atkins GJ, Riool M, Thompson K, Morgenstern M, Stoddart MJ, Richards RG, Zaat SAJ, Moriarty TF. Current Concepts of Osteomyelitis: From Pathologic Mechanisms to Advanced Research Methods. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1151-1163. [PMID: 32194053 DOI: 10.1016/j.ajpath.2020.02.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/18/2020] [Accepted: 02/27/2020] [Indexed: 01/18/2023]
Abstract
Osteomyelitis is an inflammation of the bone and bone marrow that is most commonly caused by a Staphylococcus aureus infection. Much of our understanding of the underlying pathophysiology of osteomyelitis, from the perspective of both host and pathogen, has been revised in recent years, with notable discoveries including the role played by osteocytes in the recruitment of immune cells, the invasion and persistence of S. aureus in submicron channels of cortical bone, and the diagnostic role of polymorphonuclear cells in implant-associated osteomyelitis. Advanced in vitro cell culture models, such as ex vivo culture models or organoids, have also been developed over the past decade, and have become widespread in many fields, including infectious diseases. These models better mimic the in vivo environment, allow the use of human cells, and can reduce our reliance on animals in osteomyelitis research. In this review, we provide an overview of the main pathologic concepts in osteomyelitis, with a focus on the new discoveries in recent years. Furthermore, we outline the value of modern in vitro cell culture techniques, with a focus on their current application to infectious diseases and osteomyelitis in particular.
Collapse
Affiliation(s)
- Marloes I Hofstee
- AO Research Institute Davos, Davos, Switzerland; Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research and Department of Orthopaedics, University of Rochester Medical Center, Rochester, New York
| | - Gerald J Atkins
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, South Australia, Australia
| | - Martijn Riool
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | | | - Mario Morgenstern
- Department of Orthopedic Surgery and Traumatology, University Hospital Basel, Basel, Switzerland
| | | | | | - Sebastian A J Zaat
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | | |
Collapse
|
29
|
Shlepotina NM, Peshikova MV, Kolesnikov OL, Shishkova YS. Modern Conceptions about the Mechanisms of Interaction Between Biofilm and Cellular Immunity Factors. ACTA ACUST UNITED AC 2020. [DOI: 10.36233/0372-9311-2020-1-83-90] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Features of the cellular immune response in the presence of a microbial biofilm are well described in the literature. Based on numerous studies, it became possible to establish a number of patterns: mature biofilms are better protected from immune factors, the effectiveness of antibiofilm strategies depends on species of the microorganisms, forming the biofilm, and, accordingly, on the composition of the biopolymer matrix. For example, rhamnolipids and alginate of Pseudomonas aeruginosa exert a significant negative effect on the function of immunocompetent cells. The bacteria of biofilms became able to turn to their advantage many of the protective reactions developed by the immune system and fixed evolutionarily, applying them for the growth and development of the microbial consortium.
Collapse
|
30
|
Stewart PS, Bjarnsholt T. Risk factors for chronic biofilm-related infection associated with implanted medical devices. Clin Microbiol Infect 2020; 26:1034-1038. [PMID: 32120041 DOI: 10.1016/j.cmi.2020.02.027] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/20/2020] [Accepted: 02/22/2020] [Indexed: 11/15/2022]
Abstract
BACKGROUND The use of implanted medical devices is associated with a small but clinically important risk of foreign body infection. A key question is: why do some patients develop chronic infection associated with an implanted device, but most do not? AIMS The literature on patient-specific risk factors for chronic infections associated with five types of implants was surveyed to glean clues about the etiology of these infections. SOURCES Data were collected from 47 articles through calendar year 2017 for five categories of device-related infections: cardiovascular implantable electronic devices (CIEDs), hernia meshes, prosthetic hip and knee joints, prosthetic shoulder joints and breast implants. CONTENT Important risk factors include immunomodulation/steroid therapy, diabetes, smoking, and renal disease/haemodialysis-findings that point to a critical role of a compromised innate immune response in determining vulnerable subpopulations. IMPLICATIONS A model of biofilm-related device infection is presented that posits defects in the innate immune response both systemically and locally, in the immediate vicinity of an abiotic biomaterial. The limitations of in vitro and animal models of chronic device-related infections are discussed in this context as are implications for research and clinical practice.
Collapse
Affiliation(s)
- P S Stewart
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA; Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, USA.
| | - T Bjarnsholt
- Costerton Biofilm Center, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
31
|
Abdul Hamid AI, Nakusi L, Givskov M, Chang YT, Marquès C, Gueirard P. A mouse ear skin model to study the dynamics of innate immune responses against Staphylococcus aureus biofilms. BMC Microbiol 2020; 20:22. [PMID: 31996131 PMCID: PMC6990489 DOI: 10.1186/s12866-019-1635-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/07/2019] [Indexed: 11/10/2022] Open
Abstract
Background Staphylococcus aureus is a human pathogen that is a common cause of nosocomial infections and infections on indwelling medical devices, mainly due to its ability to shift between the planktonic and the biofilm/sessile lifestyle. Biofilm infections present a serious problem in human medicine as they often lead to bacterial persistence and thus to chronic infections. The immune responses elicited by biofilms have been described as specific and ineffective. In the few experiments performed in vivo, the importance of neutrophils and macrophages as a first line of defence against biofilm infections was clearly established. However, the bilateral interactions between biofilms and myeloid cells remain poorly studied and analysis of the dynamic processes at the cellular level in tissues inoculated with biofilm bacteria is still an unexplored field. It is urgent, therefore, to develop biologically sound experimental approaches in vivo designed to extract specific immune signatures from the planktonic and biofilm forms of bacteria. Results We propose an in vivo transgenic mouse model, used in conjunction with intravital confocal microscopy to study the dynamics of host inflammatory responses to bacteria. Culture conditions were created to prepare calibrated inocula of fluorescent planktonic and biofilm forms of bacteria. A confocal imaging acquisition and analysis protocol was then drawn up to study the recruitment of innate immune cells in the skin of LysM-EGFP transgenic mice. Using the mouse ear pinna model, we showed that inflammatory responses to S. aureus can be quantified over time and that the dynamics of innate immune cells after injection of either the planktonic or biofilm form can be characterized. First results showed that the ability of phagocytic cells to infiltrate the injection site and their motility is not the same in planktonic and biofilm forms of bacteria despite the cells being considerably recruited in both cases. Conclusion We developed a mouse model of infection to compare the dynamics of the inflammatory responses to planktonic and biofilm bacteria at the tissue and cellular levels. The mouse ear pinna model is a powerful imaging system to analyse the mechanisms of biofilm tolerance to immune attacks.
Collapse
Affiliation(s)
- Aizat Iman Abdul Hamid
- Laboratoire Microorganismes : Génome et Environnement, UMR CNRS 6023, Université Clermont-Auvergne, Clermont Ferrand, France
| | - Laurence Nakusi
- Laboratoire Microorganismes : Génome et Environnement, UMR CNRS 6023, Université Clermont-Auvergne, Clermont Ferrand, France
| | - Mickael Givskov
- Costerton Biofilm Center, department of Immunology and Microbiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Young-Tae Chang
- Center for Self-assembly and Complexity, IBS and Department of Chemistry, POSTECH, Pohang, Republic of Korea
| | - Claire Marquès
- Laboratoire Microorganismes : Génome et Environnement, UMR CNRS 6023, Université Clermont-Auvergne, Clermont Ferrand, France
| | - Pascale Gueirard
- Laboratoire Microorganismes : Génome et Environnement, UMR CNRS 6023, Université Clermont-Auvergne, Clermont Ferrand, France.
| |
Collapse
|
32
|
Direct Microscopic Observation of Human Neutrophil-Staphylococcus aureus Interaction In Vitro Suggests a Potential Mechanism for Initiation of Biofilm Infection on an Implanted Medical Device. Infect Immun 2019; 87:IAI.00745-19. [PMID: 31548325 DOI: 10.1128/iai.00745-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 01/29/2023] Open
Abstract
The ability of human neutrophils to clear newly attached Staphylococcus aureus bacteria from a serum-coated glass surface was examined in vitro using time-lapse confocal scanning laser microscopy. Quantitative image analysis was used to measure the temporal change in bacterial biomass, neutrophil motility, and fraction of the surface area policed by neutrophils. In control experiments in which the surface was inoculated with bacteria but no neutrophils were added, prolific bacterial growth was observed. Neutrophils were able to control bacterial growth but only consistently when the neutrophil/bacterium number ratio exceeded approximately 1. When preattached bacteria were given a head start and allowed to grow for 3 h prior to neutrophil addition, neutrophils were unable to maintain control of the nascent biofilm. In these head-start experiments, aggregates of bacterial biofilm with areas of 50 μm2 or larger formed, and the growth of such aggregates continued even when multiple neutrophils attacked a cluster. These results suggest a model for the initiation of a biofilm infection in which a delay in neutrophil recruitment to an abiotic surface allows surface-attached bacteria time to grow and form aggregates that become protected from neutrophil clearance. Results from a computational model of the neutrophil-biofilm surface contest supported this conceptual model and highlighted the stochastic nature of the interaction. Additionally, we observed that both neutrophil motility and clearance of bacteria were impaired when oxygen tension was reduced to 0% or 2% O2.
Collapse
|
33
|
Hajdamowicz NH, Hull RC, Foster SJ, Condliffe AM. The Impact of Hypoxia on the Host-Pathogen Interaction between Neutrophils and Staphylococcus aureus. Int J Mol Sci 2019; 20:ijms20225561. [PMID: 31703398 PMCID: PMC6888323 DOI: 10.3390/ijms20225561] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/26/2019] [Accepted: 10/31/2019] [Indexed: 12/20/2022] Open
Abstract
Neutrophils are key to host defence, and impaired neutrophil function predisposes to infection with an array of pathogens, with Staphylococcus aureus a common and sometimes life-threatening problem in this setting. Both infiltrating immune cells and replicating bacteria consume oxygen, contributing to the profound tissue hypoxia that characterises sites of infection. Hypoxia in turn has a dramatic effect on both neutrophil bactericidal function and the properties of S. aureus, including the production of virulence factors. Hypoxia thereby shapes the host-pathogen interaction and the progression of infection, for example promoting intracellular bacterial persistence, enabling local tissue destruction with the formation of an encaging abscess capsule, and facilitating the establishment and propagation of bacterial biofilms which block the access of host immune cells. Elucidating the molecular mechanisms underlying host-pathogen interactions in the setting of hypoxia will enable better understanding of persistent and recalcitrant infections due to S. aureus and may uncover novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Natalia H Hajdamowicz
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Beech Hill Road, Sheffield S10 2TN, UK; (N.H.H.); (R.C.H.)
- Florey Institute, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK;
| | - Rebecca C Hull
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Beech Hill Road, Sheffield S10 2TN, UK; (N.H.H.); (R.C.H.)
- Florey Institute, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK;
| | - Simon J Foster
- Florey Institute, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK;
| | - Alison M Condliffe
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Beech Hill Road, Sheffield S10 2TN, UK; (N.H.H.); (R.C.H.)
- Florey Institute, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK;
- Correspondence:
| |
Collapse
|
34
|
|
35
|
Gorbet M, Sperling C, Maitz MF, Siedlecki CA, Werner C, Sefton MV. The blood compatibility challenge. Part 3: Material associated activation of blood cascades and cells. Acta Biomater 2019; 94:25-32. [PMID: 31226478 DOI: 10.1016/j.actbio.2019.06.020] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/03/2019] [Accepted: 06/13/2019] [Indexed: 01/09/2023]
Abstract
Following protein adsorption/activation which is the first step after the contact of material surfaces and whole blood (part 2), fibrinogen is converted to fibrin and platelets become activated and assembled in the form of a thrombus. This thrombus formation is the key feature that needs to be minimized in the creation of materials with low thrombogenicity. Further aspects of blood compatibility that are important on their own are complement and leukocyte activation which are also important drivers of thrombus formation. Hence this review summarizes the state of knowledge on all of these cascades and cells and their interactions. For each cascade or cell type, the chapter distinguishes statements which are in widespread agreement from statements where there is less of a consensus. STATEMENT OF SIGNIFICANCE: This paper is part 3 of a series of 4 reviews discussing the problem of biomaterial associated thrombogenicity. The objective was to highlight features of broad agreement and provide commentary on those aspects of the problem that were subject to dispute. We hope that future investigators will update these reviews as new scholarship resolves the uncertainties of today.
Collapse
Affiliation(s)
- Maud Gorbet
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Claudia Sperling
- Institute Biofunctional Polymer Materials, Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
| | - Manfred F Maitz
- Institute Biofunctional Polymer Materials, Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
| | - Christopher A Siedlecki
- Departments of Surgery and Bioengineering, The Pennsylvania State University, College of Medicine, Hershey, PA 17033, United States
| | - Carsten Werner
- Institute Biofunctional Polymer Materials, Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V., Dresden, Germany
| | - Michael V Sefton
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
36
|
Seebach E, Kubatzky KF. Chronic Implant-Related Bone Infections-Can Immune Modulation be a Therapeutic Strategy? Front Immunol 2019; 10:1724. [PMID: 31396229 PMCID: PMC6664079 DOI: 10.3389/fimmu.2019.01724] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/09/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic implant-related bone infections are a major problem in orthopedic and trauma-related surgery with severe consequences for the affected patients. As antibiotic resistance increases in general and because most antibiotics have poor effectiveness against biofilm-embedded bacteria in particular, there is a need for alternative and innovative treatment approaches. Recently, the immune system has moved into focus as the key player in infection defense and bone homeostasis, and the targeted modulation of the host response is becoming an emerging field of interest. The aim of this review was to summarize the current knowledge of impaired endogenous defense mechanisms that are unable to prevent chronicity of bone infections associated with a prosthetic or osteosynthetic device. The presence of foreign material adversely affects the immune system by generating a local immune-compromised environment where spontaneous clearance of planktonic bacteria does not take place. Furthermore, the surface structure of the implant facilitates the transition of bacteria from the planktonic to the biofilm stage. Biofilm formation on the implant surface is closely linked to the development of a chronic infection, and a misled adaption of the immune system makes it impossible to effectively eliminate biofilm infections. The interaction between the immune system and bone cells, especially osteoclasts, is extensively studied in the field of osteoimmunology and this crosstalk further aggravates the course of bone infection by shifting bone homeostasis in favor of bone resorption. T cells play a major role in various chronic diseases and in this review a special focus was therefore set on what is known about an ineffective T cell response. Myeloid-derived suppressor cells (MDSCs), anti-inflammatory macrophages, regulatory T cells (Tregs) as well as osteoclasts all suppress immune defense mechanisms and negatively regulate T cell-mediated immunity. Thus, these cells are considered to be potential targets for immune therapy. The success of immune checkpoint inhibition in cancer treatment encourages the transfer of such immunological approaches into treatment strategies of other chronic diseases. Here, we discuss whether immune modulation can be a therapeutic tool for the treatment of chronic implant-related bone infections.
Collapse
Affiliation(s)
- Elisabeth Seebach
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Katharina F Kubatzky
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
37
|
Josse J, Valour F, Maali Y, Diot A, Batailler C, Ferry T, Laurent F. Interaction Between Staphylococcal Biofilm and Bone: How Does the Presence of Biofilm Promote Prosthesis Loosening? Front Microbiol 2019; 10:1602. [PMID: 31379772 PMCID: PMC6653651 DOI: 10.3389/fmicb.2019.01602] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 06/26/2019] [Indexed: 12/19/2022] Open
Abstract
With the aging of population, the number of indications for total joint replacement is continuously increasing. However, prosthesis loosening can happen and is related to two major mechanisms: (1) aseptic loosening due to prosthesis micromotion and/or corrosion and release of wear particles from the different components of the implanted material and (2) septic loosening due to chronic prosthetic joint infection (PJI). The “aseptic” character of prosthesis loosening has been challenged over the years, especially considering that bacteria can persist in biofilms and be overlooked during diagnosis. Histological studies on periprosthetic tissue samples reported that macrophages are the principle cells associated with aseptic loosening due to wear debris. They produce cytokines and favor an inflammatory environment that induces formation and activation of osteoclasts, leading to bone resorption and periprosthetic osteolysis. In PJIs, the presence of infiltrates of polymorphonuclear neutrophils is a major criterion for histological diagnosis. Neutrophils are colocalized with osteoclasts and zones of osteolysis. A similar inflammatory environment also develops, leading to bone resorption through osteoclasts. Staphylococcus aureus, Staphylococcus epidermidis, and Staphylococcus lugdunensis are the main staphylococci observed in PJIs. They share the common feature to form biofilm. For S. aureus and S. epidermidis, the interaction between biofilm and immunes cells (macrophages and polymorphonuclear neutrophils) differs regarding the species. Indeed, the composition of extracellular matrix of biofilm seems to impact the interaction with immune cells. Recent papers also reported the major role of myeloid-derived suppressor cells in biofilm-associated PJIs with S. aureus. These cells prevent lymphocyte infiltration and facilitate biofilm persistence. Moreover, the role of T lymphocytes is still unclear and potentially underestimates. In this review, after introducing the cellular mechanism of aseptic and septic loosening, we will focus on the interrelationships between staphylococcal biofilm, immune cells, and bone cells.
Collapse
Affiliation(s)
- Jérôme Josse
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
| | - Florent Valour
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Yousef Maali
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Alan Diot
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Cécile Batailler
- Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Maladies Infectieuses, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Tristan Ferry
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Frédéric Laurent
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Laboratoire de Bactériologie, Institut des Agents Infectieux, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
38
|
Novel Assay To Characterize Neutrophil Responses to Oral Biofilms. Infect Immun 2019; 87:IAI.00790-18. [PMID: 30455195 DOI: 10.1128/iai.00790-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/12/2018] [Indexed: 12/25/2022] Open
Abstract
Neutrophils, the most numerous leukocytes, play an important role in maintaining oral health through interactions with oral microbial biofilms. Both neutrophil hyperactivity and the bacterial subversion of neutrophil responses can cause inflammation-mediated tissue damage like that seen in periodontal disease. We describe here an assay that assesses neutrophil activation responses to monospecies biofilm bacteria in vitro based on the surface expression of cluster of differentiation (CD) markers associated with various neutrophil functions. Most of what we know about neutrophil responses to bacteria is based on in vitro assays that use planktonic bacteria and isolated/preactivated neutrophils, which makes interpretation of the neutrophil responses to bacteria a challenge. An understanding of how neutrophils differentially interact with and respond to commensal and pathogenic oral bacteria is necessary in order to further understand the neutrophil's role in maintaining oral health and the pathogenesis of periodontal disease. In this study, a flow cytometry-based in vitro assay was developed to characterize neutrophil activation states based on CD marker expressions in response to oral monospecies bacterial biofilms. Using this approach, changes in CD marker expressions in response to specific prominent oral commensal and pathogenic bacteria were assayed. Several functional assays, including assays for phagocytosis, production of reactive oxygen species, activation of the transcription factor Nrf2, neutrophil extracellular trap formation, and myeloperoxidase release, were also performed to correlate neutrophil function with CD marker expression. Our results demonstrate that neutrophils display bacterial species-specific responses. This assay can be used to characterize how specific biofilms alter specific neutrophil pathways associated with their activation.
Collapse
|
39
|
Raafat D, Otto M, Reppschläger K, Iqbal J, Holtfreter S. Fighting Staphylococcus aureus Biofilms with Monoclonal Antibodies. Trends Microbiol 2019; 27:303-322. [PMID: 30665698 DOI: 10.1016/j.tim.2018.12.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/10/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus (S. aureus) is a notorious pathogen and one of the most frequent causes of biofilm-related infections. The treatment of S. aureus biofilms is hampered by the ability of the biofilm structure to shield bacteria from antibiotics as well as the host's immune system. Therefore, new preventive and/or therapeutic interventions, including the use of antibody-based approaches, are urgently required. In this review, we describe the mechanisms by which anti-S. aureus antibodies can help in combating biofilms, including an up-to-date overview of monoclonal antibodies currently in clinical trials. Moreover, we highlight ongoing efforts in passive vaccination against S. aureus biofilm infections, with special emphasis on promising targets, and finally indicate the direction into which future research could be heading.
Collapse
Affiliation(s)
- Dina Raafat
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Egypt; Current affiliation: Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Kevin Reppschläger
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Jawad Iqbal
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Silva Holtfreter
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
40
|
Suresh MK, Biswas R, Biswas L. An update on recent developments in the prevention and treatment of Staphylococcus aureus biofilms. Int J Med Microbiol 2019; 309:1-12. [DOI: 10.1016/j.ijmm.2018.11.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 11/19/2018] [Accepted: 11/26/2018] [Indexed: 12/17/2022] Open
|
41
|
In vitro and ex vivo systems at the forefront of infection modeling and drug discovery. Biomaterials 2018; 198:228-249. [PMID: 30384974 PMCID: PMC7172914 DOI: 10.1016/j.biomaterials.2018.10.030] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 10/05/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022]
Abstract
Bacterial infections and antibiotic resistant bacteria have become a growing problem over the past decade. As a result, the Centers for Disease Control predict more deaths resulting from microorganisms than all cancers combined by 2050. Currently, many traditional models used to study bacterial infections fail to precisely replicate the in vivo bacterial environment. These models often fail to incorporate fluid flow, bio-mechanical cues, intercellular interactions, host-bacteria interactions, and even the simple inclusion of relevant physiological proteins in culture media. As a result of these inadequate models, there is often a poor correlation between in vitro and in vivo assays, limiting therapeutic potential. Thus, the urgency to establish in vitro and ex vivo systems to investigate the mechanisms underlying bacterial infections and to discover new-age therapeutics against bacterial infections is dire. In this review, we present an update of current in vitro and ex vivo models that are comprehensively changing the landscape of traditional microbiology assays. Further, we provide a comparative analysis of previous research on various established organ-disease models. Lastly, we provide insight on future techniques that may more accurately test new formulations to meet the growing demand of antibiotic resistant bacterial infections.
Collapse
|
42
|
Wu X, Zhang Y, Chen X, Chen J, Jia M. Inflammatory immune response in rabbits with Staphylococcus aureus biofilm-associated sinusitis. Int Forum Allergy Rhinol 2018; 8:1226-1232. [PMID: 29979838 PMCID: PMC6282565 DOI: 10.1002/alr.22175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 05/21/2018] [Accepted: 06/08/2018] [Indexed: 12/19/2022]
Abstract
Background Staphylococcus aureus is the most commonly isolated bacterium from patients with surgically recalcitrant chronic rhinosinusitis (CRS). Understanding the immune responses to S aureus biofilms will provide insights into how the host response may be manipulated by therapeutic agents to improve the chances of successfully preventing and treating these infections. In this study, we investigated the inflammatory immune response in a rabbit model of S aureus biofilm–related sinusitis by analyzing the levels of some major inflammatory cytokines. Methods Eighteen New Zealand white rabbits were randomly divided into 3 groups: a blank‐control group; a negative‐control group; and a model group. Four weeks after the biofilm‐associated sinusitis models were established, the sinus mucosa was harvested and examined using hematoxylin‐eosin (H&E) staining, scanning electron microscopy (SEM), reverse transcription polymerase chain reaction (RT‐PCR), and western blotting. The expression levels of inflammatory cytokines were analyzed statistically. Results Interleukin (IL)‐1β, IL‐8, and tumor necrosis factor (TNF)‐α expression levels were significantly higher in the model group than in the blank‐control group (p < 0.05); mRNA levels were increased by 1600%, 230%, and 130%, respectively, and the protein levels were increased by 180%, 100%, and 100%, respectively. In contrast, IL‐4 and IL‐5 mRNA levels were reduced by 44% and 70%, respectively, compared with the blank‐control group (p < 0.05). Conclusion S aureus biofilms in the rabbit maxillary sinus mucosa were associated with increased IL‐1β, IL‐8, and TNF‐α expression, and decreased IL‐4 and IL‐5 expression.
Collapse
Affiliation(s)
- Xianmin Wu
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yue Zhang
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyun Chen
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jun Chen
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Minghui Jia
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
43
|
Capote-Bonato F, Bonato DV, Ayer IM, Magalhães LF, Magalhães GM, Pereira da Câmara Barros FF, Teixeira PPM, Crivellenti LZ, Negri M, Svidzinski TIE. Murine model for the evaluation of candiduria caused by Candida tropicalis from biofilm. Microb Pathog 2018; 117:170-174. [PMID: 29471135 DOI: 10.1016/j.micpath.2018.02.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/10/2017] [Accepted: 02/17/2018] [Indexed: 12/31/2022]
Abstract
To evaluate the pathophysiology of catheter-associated candiduria, the bladders of female mice were infected with Candida tropicalis. One group was implanted with a catheter fragment with preformed biofilm by cystotomy technique, while another group received, in separate, a sterile catheter fragment and a correspondent yeast suspension. The bladder tissues were examined by histopathology and the quantity of colony forming units was evaluated. All the animals presented inflammation and the presence of C. tropicalis was observed in the tissue within 72 h of the introduction of biofilm, while 75% of the mice remained infected after 144 h. However, only 50% of animals from the group infected with C. tropicalis in suspension (planktonic yeasts), exhibited such signs of infection over time. The cystotomy technique is therefore viable in mice, and is an effective model for evaluating the pathogenesis of candiduria from catheter biofilms. The model revealed the potential of C. tropicalis infectivity and demonstrated more effective evasion of the host response in biofilm form than the planktonic yeast.
Collapse
Affiliation(s)
| | | | - Ilan Munhoz Ayer
- Department of Animal Science (DAS), Franca University, São Paulo, Brazil
| | | | | | | | | | | | - Melyssa Negri
- Department of Clinical Analysis (DCA), State University of Maringá, Paraná, Brazil
| | | |
Collapse
|
44
|
Meyle E, Stroh P, GüNther F, Hoppy-Tichy T, Wagner C, HäNsch GM. Destruction of Bacterial Biofilms by Polymorphonuclear Neutrophils: Relative contribution of Phagocytosis, DNA Release, and Degranulation. Int J Artif Organs 2018; 33:608-20. [DOI: 10.1177/039139881003300906] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2010] [Indexed: 12/13/2022]
Abstract
Bacteria organized in biofilms are a common cause of relapsing or persistent infections, and the ultimate cause of implant-associated osteomyelitis. In these patients, biofilms of staphylococci are prevalent. Bacteria organized as biofilms are relatively resistant towards antibiotics and biocides, and it is also assumed that they may escape host defense mechanisms. In this context, we have studied how polymorphonuclear neutrophils (PMN), the “first line of defense” against bacterial infection, interact with biofilms generated in vitro. We found that PMN recognize biofilms and activate defense-associated reactions, including phagocytosis, degranulation of lactoferrin and elastase, and DNA release as well. Destruction of biofilms ensues, showing that biofilms are not inherently protected against the attack by phagocytic cells.
Collapse
Affiliation(s)
- Eva Meyle
- Institute of Immunology, University of Heidelberg, Heidelberg - Germany
- Institute of Pharmacy, University of Heidelberg, Heidelberg - Germany
| | - Petra Stroh
- Institute of Immunology, University of Heidelberg, Heidelberg - Germany
| | - Frank GüNther
- Institute of Immunology, University of Heidelberg, Heidelberg - Germany
| | | | - Christof Wagner
- Department of Trauma Surgery and Orthopedic Surgery, BG Trauma Clinic Ludwigshafen, Ludwigshafen - Germany
| | | |
Collapse
|
45
|
Abstract
Microbial biofilms can colonize medical devices and human tissues, and their role in microbial pathogenesis is now well established. Not only are biofilms ubiquitous in natural and human-made environments, but they are also estimated to be associated with approximately two-thirds of nosocomial infections. This multicellular aggregated form of microbial growth confers a remarkable resistance to killing by antimicrobials and host defenses, leading biofilms to cause a wide range of subacute or chronic infections that are difficult to eradicate. We have gained tremendous knowledge on the molecular, genetic, microbiological, and biophysical processes involved in biofilm formation. These insights now shape our understanding, diagnosis, and management of many infectious diseases and direct the development of novel antimicrobial therapies that target biofilms. Bacterial and fungal biofilms play an important role in a range of diseases in pulmonary and critical care medicine, most importantly catheter-associated infections, ventilator-associated pneumonia, chronic Pseudomonas aeruginosa infections in cystic fibrosis lung disease, and Aspergillus fumigatus pulmonary infections.
Collapse
|
46
|
Lin HF, Liao KF, Chang CM, Lin CL, Lai SW. Statin use correlates with reduced risk of chronic osteomyelitis: a nationwide case-control study in Taiwan. Curr Med Res Opin 2017; 33:2235-2240. [PMID: 28699801 DOI: 10.1080/03007995.2017.1354831] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVE Potential association between prior statin use and chronic osteomyelitis is examined. METHODS A nationwide case-control study was conducted based on data taken from the Taiwan National Health Insurance program. The case group includes 2338 subjects aged 20-84 years newly diagnosed for chronic osteomyelitis from 2000 to 2013; the control group included 2338 randomly selected subjects without chronic osteomyelitis matched for sex, age, and index year. Statin use was respectively defined as "current", "recent" or "past" if the most recent statin prescription was filled <3 months, 3-6 months or ≥6 months prior to the chronic osteomyelitis diagnosis. Relative risk of chronic osteomyelitis associated with statin use was measured by the odds ratio (OR) with 95% confidence interval (CI) using the conditional logistic regression model. RESULTS After controlling for potential confounders, the adjusted ORs of chronic osteomyelitis were 0.57 for subjects with current statin use (95% CI 0.45, 0.72), 0.80 for subjects with recent statin use (95% CI 0.48, 1.33), and 1.00 for subjects with past statin use (95% CI 0.83, 1.20), compared patients with no prior statin use. In further analysis, the adjusted ORs of chronic osteomyelitis were 0.70 for subjects with cumulative statin use <12 months (95% CI 0.47, 1.07), and 0.56 for subjects with cumulative statins use ≥12 months (95% CI 0.41, 0.77), compared with those with no prior statin use. CONCLUSIONS Current statin use is associated with reduced concurrent diagnosis of chronic osteomyelitis, particularly for a cumulative statin use ≥12 months.
Collapse
Affiliation(s)
- Hsien-Feng Lin
- a School of Chinese Medicine , China Medical University , Taichung , Taiwan
- b Department of Family Medicine , China Medical University Hospital , Taichung , Taiwan
| | - Kuan-Fu Liao
- c Department of Internal Medicine , Taichung Tzu Chi General Hospital , Taichung , Taiwan
- d College of Medicine , Tzu Chi University , Hualien , Taiwan
- e Graduate Institute of Integrated Medicine, China Medical University , Taichung , Taiwan
| | - Ching-Mei Chang
- f Department of Nursing , Tungs' Taichung Metro Habor Hospital , Taichung , Taiwan
| | - Cheng-Li Lin
- g College of Medicine , China Medical University , Taichung , Taiwan
- h Management Office for Health Data, China Medical University Hospital , Taichung , Taiwan
| | - Shih-Wei Lai
- b Department of Family Medicine , China Medical University Hospital , Taichung , Taiwan
- g College of Medicine , China Medical University , Taichung , Taiwan
| |
Collapse
|
47
|
Biofilms: Survival and defense strategy for pathogens. Int J Med Microbiol 2017; 307:481-489. [PMID: 28950999 DOI: 10.1016/j.ijmm.2017.09.016] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 09/11/2017] [Accepted: 09/19/2017] [Indexed: 01/20/2023] Open
Abstract
Studies on biofilm related infections are gaining prominence owing to their involvement in majority of clinical infections. Biofilm, considered as a generic mechanism for survival used by pathogenic as well as non-pathogenic microorganisms, involves surface attachment and growth of heterogeneous cells encapsulated within a matrix. The matrix provides ecological niche where partnership of cells endows a community like behaviour that not only enables the cohort to survive local microenvironment stress but also channelizes them to evolve, disseminate and cause resurgence of infections. In this mini-review we highlight the mechanisms used by microbes to develop and sustain biofilms, including the influence of the microbiota. Several strategies to target biofilms have been validated on certain groups of microorganisms and these basically target different stages in the life cycle of biofilm, however comprehensive methods to target microbial biofilms are relatively unknown. In the backdrop of recent reports suggesting that biofilms can harbour multiple species of organisms, we need to relook and devise newer strategies against biofilms. Effective anti-biofilm strategies cannot be confined to a single methodology that can disrupt one pathway but should simultaneously target the various routes adopted by the microorganisms for survival within their ecosystem. An overview of the currently available drugs, their mode of action, genomic targets and translational therapies against biofilm related infection are discussed.
Collapse
|
48
|
Lockhart JS, Buret AG, Ceri H, Storey DG, Anderson SJ, Morck DW. Mixed species biofilms of Fusobacterium necrophorum and Porphyromonas levii impair the oxidative response of bovine neutrophils in vitro. Anaerobe 2017; 47:157-164. [PMID: 28526497 DOI: 10.1016/j.anaerobe.2017.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 05/08/2017] [Accepted: 05/15/2017] [Indexed: 01/11/2023]
Abstract
Biofilms composed of anaerobic bacteria can result in persistent infections and chronic inflammation. Host immune cells have difficulties clearing biofilm-related infections and this can result in tissue damage. Neutrophils are a vital component of the innate immune system and help clear biofilms. The comparative neutrophilic response to biofilms versus planktonic bacteria remains incompletely understood, particularly in the context of mixed infections. The objective of this study was to generate mixed species anaerobic bacterial biofilms composed of two opportunistic pathogens, Fusobacterium necrophorum and Porphyromonas levii, and evaluate neutrophil responses to extracellular fractions from both biofilms and planktonic cell co-cultures of the same bacteria. Purified bovine neutrophils exposed to culture supernatants from mixed species planktonic bacteria showed elevated oxidative activity compared to neutrophils exposed to biofilms composed of the same bacteria. Bacterial lipopolysaccharide plays a significant role in the stimulation of neutrophils; biofilms produced substantially more lipopolysaccharide than planktonic bacteria under these experimental conditions. Removal of lipopolysaccharide significantly reduced neutrophil oxidative response to culture supernatants of planktonic bacteria. Oxidative responses to LPS-removed biofilm supernatants and LPS-removed planktonic cell supernatants were similar. The limited neutrophil response to biofilm bacteria observed in this study supports the reduced ability of the innate immune system to eradicate biofilm-associated infections. Lipopolysaccharide is likely important in neutrophil response; however, the presence of other extracellular, immune modifying molecules in the bacterial media also appears to be important in altering neutrophil function.
Collapse
Affiliation(s)
- Joey S Lockhart
- Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada; Biofilm Research Group, University of Calgary, Calgary, Alberta T2N 1N4, Canada.
| | - Andre G Buret
- Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada; Biofilm Research Group, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Howard Ceri
- Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada; Biofilm Research Group, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Douglas G Storey
- Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada; Biofilm Research Group, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | | | - Douglas W Morck
- Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada; Biofilm Research Group, University of Calgary, Calgary, Alberta T2N 1N4, Canada; Animal Health Unit, University of Calgary, Calgary, Alberta T2N 1N4, Canada; Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada.
| |
Collapse
|
49
|
Günther F, Blessing B, Tacconelli E, Mutters NT. MRSA decolonization failure-are biofilms the missing link? Antimicrob Resist Infect Control 2017; 6:32. [PMID: 28360994 PMCID: PMC5371339 DOI: 10.1186/s13756-017-0192-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/21/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Device-associated infections due to biofilm-producing methicillin-resistant Staphylococcus aureus (MRSA) have been recently associated with the failure of antibiotic treatment and decolonization measures. The goal of our study was to evaluate the extent to which the formation of biofilms influenced the efficacy of topical decolonization agents or disinfectants such as mupirocin (MUP), octenidine (OCT), chlorhexidine (CHG), polyhexanide (POL), and chloroxylenol (CLO). METHODS Bacterial killing in biofilms by the disinfectants and MUP was determined as the reduction [%] in metabolic activity determined by a biofilm viability assay that uses kinetic analysis of metabolic activity. The test substances were diluted in water with standardized hardness (WSH) at 25 °C at the standard concentration as well as half the standard concentration to demonstrate the dilution effects in a practical setting. The tested concentrations were: CHG 1%, 2%; OCT 0.1%, 0.05%; PH 0.04%, 0.02%; and CLO 0.12%, 0.24%. A test organism suspension, 1 mL containing ~1 × 109 bacterial cells/mL, and 1 mL of sterile WSH were mixed and incubated for six different exposure times (15 s, 1, 3, 5, 10 and 20 min) after the test substance was added. Additionally, the bactericidal effects of all substances were tested on planktonic bacteria and measured as the log10 reduction. RESULTS The disinfectants OCT and CHG showed good efficacy in inhibiting MRSA in biofilms with reduction rates of 94 ± 1% and 91 ± 1%, respectively. POL, on the other hand, had a maximum efficacy of only 81 ± 7%. Compared to the tested disinfectants, MUP showed a significantly lower efficacy with <20% inhibition (p < .05). Bactericidal effects were the greatest for CHG (log10 reduction of 9.0), followed by OCT (7.7), POL (5.1), and CLO (6.8). MUP, however, showed a very low bactericidal effect of only 2.1. Even when the exposure time was increased to 24 h, 2% MUP did not show sufficient bactericidal effect. CONCLUSIONS Our data provide evidence that OCT and CHG are effective components for disinfection of MRSA-biofilms. On the other hand, exposure to MUP at the standard concentrations in topical preparations did not effectively inhibit MRSA-biofilms and also did not show adequate bactericidal effects. Combining an MUP-based decolonization regimen with a disinfectant such as OCT or CHG could decrease decolonization failure.
Collapse
Affiliation(s)
- Frank Günther
- Department of Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69126 Heidelberg, Germany
| | - Brigitte Blessing
- Department of Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69126 Heidelberg, Germany
| | - Evelina Tacconelli
- Division of Infectious Diseases - Department of Internal Medicine I, Tübingen University Hospital, Tübingen, Germany
- German Centre for Infection Research (DZIF), Tübingen, Germany
| | - Nico T. Mutters
- Department of Infectious Diseases, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69126 Heidelberg, Germany
- Division of Infectious Diseases - Department of Internal Medicine I, Tübingen University Hospital, Tübingen, Germany
| |
Collapse
|
50
|
Balta E, Stopp J, Castelletti L, Kirchgessner H, Samstag Y, Wabnitz GH. Qualitative and quantitative analysis of PMN/T-cell interactions by InFlow and super-resolution microscopy. Methods 2017; 112:25-38. [DOI: 10.1016/j.ymeth.2016.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 10/20/2022] Open
|