Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 458
Filter
1.
Hum Pathol ; 147: 5-14, 2024 May.
Article in English | MEDLINE | ID: mdl-38280657

ABSTRACT

Technological and implant design advances have helped reduce the frequency of aseptic total joint arthroplasty failure, but periprosthetic joint infections (PJI) remain a clinical important problem with high patient morbidity. Misinterpreting PJI as aseptic mechanical loosening commonly leads to unsatisfactory revision arthroplasty, persistent infection, and poor long-term results. While there is no single "gold standard" diagnostic test for PJI, recent collaborative efforts by Orthopaedic and Infectious Disease Societies have developed algorithms for diagnosing PJI. However, the efficacy of individual tests as well as diagnostic thresholds are controversial. We review the recommended thresholds for commonly used screening tests as well as tissue histopathology and confirmatory tests to diagnose periprosthetic infection. We also update lesser-known laboratory tests, and we briefly summarize rapidly evolving molecular tests to diagnose periprosthetic infection. Pathologists hold a critical role in assisting with PJI diagnosis, maintaining laboratory test quality and interpreting test results. Collaboration between clinicians and pathologists is essential to provide optimal patient care and reduce the burden of PJI.


Subject(s)
Prosthesis-Related Infections , Humans , Prosthesis-Related Infections/diagnosis , Prosthesis-Related Infections/pathology , Predictive Value of Tests , Arthroplasty, Replacement/adverse effects , Arthroplasty, Replacement/instrumentation
2.
Spine J ; 23(9): 1389-1399, 2023 09.
Article in English | MEDLINE | ID: mdl-37247639

ABSTRACT

BACKGROUND CONTEXT: Bacterial infection of spinal instrumentation is a significant challenge in spinal fusion surgery. Although the intraoperative local application of powdered vancomycin is common practice for mitigating infection, the antimicrobial effects of this route of administration are short-lived. Therefore, novel antibiotic-loaded bone grafts as well as a reliable animal model to permit the testing of such therapies are needed to improve the efficacy of infection reduction practices in spinal fusion surgery. PURPOSE: This study aims to establish a clinically relevant rat model of spinal implant-associated infection to permit the evaluation of antimicrobial bone graft materials used in spinal fusion. STUDY DESIGN: Rodent study of chronic spinal implant-associated infection. METHODS: Instrumentation anchored in and spanning the vertebral bodies of L4 and L5 was inoculated with bioluminescent methicillin-resistant Staphylococcus aureus bacteria (MRSA). Infection was monitored using an in vivo imaging system (IVIS) for 8 weeks. Spines were harvested and evaluated histologically, and colony-forming units (CFUs) were quantified in harvested implants and spinal tissue. RESULTS: Postsurgical analysis of bacterial infection in vivo demonstrated stratification between MRSA and phosphate-buffered saline (PBS) control groups during the first 4 weeks of the 8-week infection period, indicating the successful establishment of acute infection. Over the 8-week chronic infection period, groups inoculated with 1 × 105 MRSA CFU and 1 × 106 MRSA CFU demonstrated significantly higher bioluminescence than groups inoculated with PBS control (p = 0.009 and p = 0.041 respectively). Histological examination at 8 weeks postimplantation revealed the presence of abscesses localized to implant placement in all MRSA inoculation groups, with the most pervasive abscess formation in samples inoculated with 1 × 105 MRSA CFU and 1 × 106 MRSA CFU. Quantification of CFU plated from harvested spinal tissue at 8 weeks post-implantation revealed the 1 × 105 MRSA CFU inoculation group as the only group with a significantly greater average CFU count compared to PBS control (p = 0.017). Further, CFU quantification from harvested spinal tissue was greater than CFU quantification from harvested implants across all inoculation groups. CONCLUSION: Our model demonstrated that the inoculation dosage of 1 × 105 MRSA CFU exhibited the most robust chronic infection within instrumented vertebral bodies. This dosage had the greatest difference in bioluminescence signal from control (p < 0.01), the lowest mortality (0% compared to 50% for samples inoculated with 1 × 106 MRSA CFU), and a significantly higher amount of CFUs from harvested spine samples than CFUs from control harvested spine samples. Further, histological analysis confirmed the reliability of this novel rodent model of implanted-associated infection to establish infection and biofilm formation of MRSA for all inoculation groups. CLINICAL SIGNIFICANCE: This model is intended to simulate the infection of instrumentation used in spinal fusion surgeries concerning implant locality and material. This model may evaluate potential antimicrobial and osteogenic biomaterials and investigate the relationship between implant-associated infection and failed fusion.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Prosthesis-Related Infections , Staphylococcal Infections , Rats , Animals , Staphylococcal Infections/drug therapy , Persistent Infection , Rodentia , Reproducibility of Results , Prosthesis-Related Infections/drug therapy , Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/pathology , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal
3.
Malays J Pathol ; 43(3): 405-411, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34958062

ABSTRACT

The preoperative diagnosis of infection during joint arthroplasty is important for clinical management. However, the evaluation of polymorphonuclear leukocytes (PMNs) during frozen section analysis is sometimes difficult due to frozen artifacts. In the present study, we sought to investigate the utility of intraoperative fresh frozen section (FFS) examination for diagnosis of infection and to evaluate whether the neutrophil-specific surface marker CD66b helps to improve the diagnostic accuracy of infection. A consecutive series of 65 original frozen sections at the time of resection arthroplasty was retrospectively reviewed compared with corresponding permanent sections. The presence of PMNs was determined using intraoperative FFS and permanent sections. Furthermore, CD66b staining was performed to identify PMNs clearly. The ratio of male to female patients was 21:42. The mean age was 70 years. Postoperatively, 25 of 65 cases were histologically diagnosed with infection (25/65; 39%). The sensitivity and specificity of intraoperative FFS relative to permanent section histology were 100% (25/25) and 95% (38/40), respectively. Among 40 patients without infection, two showed false-positive results during intraoperative FFS diagnosis (2/40, 5%). In addition, on CD66b staining, six cases (9%) experienced changes in results, which altered the sensitivity and specificity of intraoperative FFS compared with permanent histology only to 87% and 87%, respectively. In conclusion, the diagnostic performance of intraoperative FFS is high and comparable to yields of permanent section histology. Therefore, intraoperative FFS is highly suitable diagnostic method for detection of infection during joint arthroplasty. And CD66b immunostaining facilitates delicate identification of PMNs, especially in equivocal cases.


Subject(s)
Frozen Sections , Prosthesis-Related Infections , Aged , Arthroplasty , Female , Humans , Male , Prosthesis-Related Infections/diagnosis , Prosthesis-Related Infections/pathology , Prosthesis-Related Infections/surgery , Reoperation/methods , Retrospective Studies , Sensitivity and Specificity , Staining and Labeling
4.
PLoS One ; 16(9): e0257471, 2021.
Article in English | MEDLINE | ID: mdl-34520499

ABSTRACT

16S rRNA gene sequencing of DNA extracted from clinically uninfected hip and knee implant samples has revealed polymicrobial populations. However, previous studies assessed 16S rRNA gene sequencing as a technique for the diagnosis of periprosthetic joint infections, leaving the microbiota of presumed aseptic hip and knee implants largely unstudied. These communities of microorganisms might play important roles in aspects of host health, such as aseptic loosening. Therefore, this study sought to characterize the bacterial composition of presumed aseptic joint implant microbiota using next generation 16S rRNA gene sequencing, and it evaluated this method for future investigations. 248 samples were collected from implants of 41 patients undergoing total hip or knee arthroplasty revision for presumed aseptic failure. DNA was extracted using two methodologies-one optimized for high throughput and the other for human samples-and amplicons of the V4 region of the 16S rRNA gene were sequenced. Sequencing data were analyzed and compared with ancillary specific PCR and microbiological culture. Computational tools (SourceTracker and decontam) were used to detect and compensate for environmental and processing contaminants. Microbial diversity of patient samples was higher than that of open-air controls and differentially abundant taxa were detected between these conditions, possibly reflecting a true microbiota that is present in clinically uninfected joint implants. However, positive control-associated artifacts and DNA extraction methodology significantly affected sequencing results. As well, sequencing failed to identify Cutibacterium acnes in most culture- and PCR-positive samples. These challenges limited characterization of bacteria in presumed aseptic implants, but genera were identified for further investigation. In all, we provide further support for the hypothesis that there is likely a microbiota present in clinically uninfected joint implants, and we show that methods other than 16S rRNA gene sequencing may be ideal for its characterization. This work has illuminated the importance of further study of microbiota of clinically uninfected joint implants with novel molecular and computational tools to further eliminate contaminants and artifacts that arise in low bacterial abundance samples.


Subject(s)
Bacteria/isolation & purification , Microbiota , Prosthesis-Related Infections/microbiology , Adult , Aged , Arthroplasty, Replacement, Hip , Arthroplasty, Replacement, Knee , Artifacts , Bacteria/genetics , Female , Hip Joint/microbiology , Humans , Knee Joint/microbiology , Male , Middle Aged , Polymerase Chain Reaction , Prosthesis-Related Infections/pathology , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Sequence Analysis, DNA
5.
Front Immunol ; 12: 651515, 2021.
Article in English | MEDLINE | ID: mdl-33815412

ABSTRACT

Staphylococcus aureus is the predominant pathogen causing osteomyelitis. Unfortunately, no immunotherapy exists to treat these very challenging and costly infections despite decades of research, and numerous vaccine failures in clinical trials. This lack of success can partially be attributed to an overreliance on murine models where the immune correlates of protection often diverge from that of humans. Moreover, S. aureus secretes numerous immunotoxins with unique tropism to human leukocytes, which compromises the targeting of immune cells in murine models. To study the response of human immune cells during chronic S. aureus bone infections, we engrafted non-obese diabetic (NOD)-scid IL2Rγnull (NSG) mice with human hematopoietic stem cells (huNSG) and analyzed protection in an established model of implant-associated osteomyelitis. The results showed that huNSG mice have increases in weight loss, osteolysis, bacterial dissemination to internal organs, and numbers of Staphylococcal abscess communities (SACs), during the establishment of implant-associated MRSA osteomyelitis compared to NSG controls (p < 0.05). Flow cytometry and immunohistochemistry demonstrated greater human T cell numbers in infected versus uninfected huNSG mice (p < 0.05), and that T-bet+ human T cells clustered around the SACs, suggesting S. aureus-mediated activation and proliferation of human T cells in the infected bone. Collectively, these proof-of-concept studies underscore the utility of huNSG mice for studying an aggressive form of S. aureus osteomyelitis, which is more akin to that seen in humans. We have also established an experimental system to investigate the contribution of specific human T cells in controlling S. aureus infection and dissemination.


Subject(s)
Abscess/immunology , Osteolysis/immunology , Osteomyelitis/immunology , Prosthesis-Related Infections/immunology , Staphylococcal Infections/immunology , Abscess/microbiology , Abscess/pathology , Animals , Disease Models, Animal , Female , Hematopoietic Stem Cell Transplantation , Humans , Mice , Osteolysis/microbiology , Osteolysis/pathology , Osteomyelitis/microbiology , Osteomyelitis/pathology , Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/pathology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/immunology , Transplantation Chimera/immunology
6.
Int J Infect Dis ; 106: 302-307, 2021 May.
Article in English | MEDLINE | ID: mdl-33819605

ABSTRACT

OBJECTIVES: In the microbiological diagnosis of periprosthetic joint infection (PJI), there is much discussion about the methodology of obtaining proper specimens, the processing technique, and suitable culture media. This retrospective study was conducted to analyse the accuracy of our culture techniques. METHODS: Tissue samples and components from 258 patients after revision arthroplasty of the hip, knee, and shoulder were investigated, and the results of tissue cultures (TC) were compared to those of sonicate fluid cultures (SFC). Furthermore, an evaluation was performed of the influence of different culture media on the detection rate. RESULTS: PJI was confirmed in 186 patients. The overall sensitivity of TC was no different to that of SFC (91.3% vs 90.8%, P = 1). In 153 cases (82.3%), TC and SFC showed concordant positive results. Results were discordant in 33 cases (17.7%). When differentiated according to the type of infection, TC showed significantly better results than SFC in detecting polymicrobial infections (97.0% vs 67.0%, P = 0.004). There were also significant differences between the culture media regarding the yield of microorganisms. CONCLUSIONS: TC was more effective in detecting co-infections. The best results were obtained using both TC and SFC. The choice of culture media has a significant influence on the quality of results.


Subject(s)
Arthritis, Infectious/diagnosis , Prostheses and Implants/microbiology , Prosthesis-Related Infections/diagnosis , Sonication , Adult , Aged , Aged, 80 and over , Arthritis, Infectious/pathology , Arthroplasty/adverse effects , Humans , Male , Middle Aged , Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/pathology , Retrospective Studies
8.
Sci Rep ; 11(1): 1622, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33452271

ABSTRACT

Implant-associated infections are challenging to diagnose and treat. Fluorescent probes have been heralded as a technologic advancement that can improve our ability to non-invasively identify infecting organisms, as well as guide the inexact procedure of surgical debridement. This study's purpose was to compare two fluorescent probes for their ability to localize Staphylococcus aureus biofilm infections on spinal implants utilizing noninvasive optical imaging, then assessing the broader applicability of the more successful probe in other infection animal models. This was followed by real-time, fluorescence image-guided surgery to facilitate debridement of infected tissue. The two probe candidates, a labelled antibiotic that targets peptidoglycan (Vanco-800CW), and the other, a labelled antibody targeting the immunodominant Staphylococcal antigen A (1D9-680), were injected into mice with spine implant infections. Mice were then imaged noninvasively with near infrared fluorescent imaging at wavelengths corresponding to the two probe candidates. Both probes localized to the infection, with the 1D9-680 probe showing greater fidelity over time. The 1D9-680 probe was then tested in mouse models of shoulder implant and allograft infection, demonstrating its broader applicability. Finally, an image-guided surgery system which superimposes fluorescent signals over analog, real-time, tissue images was employed to facilitate debridement of fluorescent-labelled bacteria.


Subject(s)
Biofilms/growth & development , Fluorescent Dyes/chemistry , Optical Imaging/methods , Prosthesis-Related Infections/surgery , Spinal Cord/diagnostic imaging , Staphylococcus aureus/isolation & purification , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Disease Models, Animal , Mice , Prostheses and Implants , Prosthesis-Related Infections/drug therapy , Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/pathology , Spinal Cord/surgery , Staphylococcus aureus/physiology , Surgery, Computer-Assisted , Tomography, X-Ray Computed , Vancomycin/pharmacology , Vancomycin/therapeutic use
9.
Int J Med Sci ; 18(2): 295-303, 2021.
Article in English | MEDLINE | ID: mdl-33390798

ABSTRACT

Background: Implant loosening - either infectious or aseptic- is a still a major complication in the field of orthopaedic surgery. In both cases, a pro-inflammatory peri-prosthetic environment is generated by the immune system - either triggered by bacteria or by implant wear particles - which leads to osteoclast differentiation and osteolysis. Since infectious cases in particular often require multiple revision surgeries, we wondered whether commonly used surgical suture material may also activate the immune system and thus contribute to loss of bone substance by generation of osteoclasts. Methods: Tissue samples from patients suffering from infectious implant loosening were collected intraoperatively and presence of osteoclasts was evaluated by histopathology and immunohistochemistry. Further on, human monocytes were isolated from peripheral blood and stimulated with surgical suture material. Cell supernatant samples were collected and ELISA analysis for the pro-inflammatory cytokine IL-8 was performed. These experiments were additionally carried out on ivory slices to demonstrate functionality of osteoclasts. Whole blood samples were incubated with surgical suture material and up-regulation of activation-associated cell surface markers CD11b and CD66b on neutrophils was evaluated by flow cytofluorometry analysis. Results: We were able to demonstrate that multinucleated giant cells form in direct vicinity to surgical suture material. These cells stained positive for cathepsin K, which is a typical protease found in osteoclasts. By in vitro analysis, we were able to show that monocytes differentiated into osteoclasts when stimulated with surgical suture material. Resorption pits on ivory slices provided proof that the osteoclasts were functional. Release of IL-8 into cell supernatant was increased after stimulation with suture material and was further enhanced if minor amounts of bacterial lipoteichoic acid (LTA) were added. Neutrophils were also activated by surgical suture material and up-regulation of CD11b and CD66b could be seen. Conclusion: We were able to demonstrate that surgical suture material induces a pro-inflammatory response of immune cells which leads to osteoclast differentiation, in particular in combination with bacterial infection. In conclusion, surgical suture material -aside from bacteria and implant wear particles- is a contributing factor in implant loosening.


Subject(s)
Orthopedic Procedures/adverse effects , Osteolysis/immunology , Prostheses and Implants/adverse effects , Prosthesis-Related Infections/immunology , Sutures/adverse effects , Adult , Aged , Aged, 80 and over , Cell Differentiation/immunology , Female , Humans , Male , Middle Aged , Orthopedic Procedures/instrumentation , Orthopedic Procedures/methods , Osteoclasts/pathology , Osteolysis/prevention & control , Prosthesis Failure , Prosthesis-Related Infections/pathology
10.
J Orthop Res ; 39(2): 389-401, 2021 02.
Article in English | MEDLINE | ID: mdl-33336806

ABSTRACT

Osteomyelitis is a devastating complication of orthopaedic surgery and commonly caused by Staphylococcus aureus (S. aureus) and Group B Streptococcus (GBS, S. agalactiae). Clinically, S. aureus osteomyelitis is associated with local inflammation, abscesses, aggressive osteolysis, and septic implant loosening. In contrast, S. agalactiae orthopaedic infections generally involve soft tissue, with acute life-threatening vascular spread. While preclinical models that recapitulate the clinical features of S. aureus bone infection have proven useful for research, no animal models of S. agalactiae osteomyelitis exist. Here, we compared the pathology caused by these bacteria in an established murine model of implant-associated osteomyelitis. In vitro scanning electron microscopy and CFU quantification confirmed similar implant inocula for both pathogens (~105 CFU/pin). Assessment of mice at 14 days post-infection demonstrated increased S. aureus virulence, as S. agalactiae infected mice had significantly greater body weight, and fewer CFU on the implant and in bone and adjacent soft tissue (p < 0.05). X-ray, µCT, and histologic analyses showed that S. agalactiae induced significantly less osteolysis and implant loosening, and fewer large TRAP+ osteoclasts than S. aureus without inducing intraosseous abscess formation. Most notably, transmission electron microscopy revealed that although both bacteria are capable of digesting cortical bone, S. agalactiae have a predilection for colonizing blood vessels embedded within cortical bone while S. aureus primarily colonizes the osteocyte lacuno-canalicular network. This study establishes the first quantitative animal model of S. agalactiae osteomyelitis, and demonstrates a vasculotropic mode of S. agalactiae infection, in contrast to the osteotropic behavior of S. aureus osteomyelitis.


Subject(s)
Bone and Bones/ultrastructure , Host-Pathogen Interactions , Osteomyelitis/microbiology , Staphylococcus aureus/physiology , Streptococcus agalactiae/physiology , Animals , Bone and Bones/microbiology , Mice , Osteomyelitis/pathology , Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/pathology , Staphylococcal Infections/pathology , Streptococcal Infections/pathology
11.
Biomed Res Int ; 2020: 3704285, 2020.
Article in English | MEDLINE | ID: mdl-33294439

ABSTRACT

Periprosthetic joint infection (PJI) is a devastating complication after arthroplasty. Prompt establishment of an infection diagnosis is critical but can be very challenging at present. In order to evaluate the diagnostic accuracy of alpha-defensin or leukocyte esterase for PJI, we performed systematic research in PubMed, Embase, and Cochrane Library to retrieve relevant studies. Data extraction and quality assessment were performed by two reviewers independently. A total of thirty-one eligible studies were finally included in the quantitative analysis. The pooled sensitivity and specificity of alpha-defensin (21 studies) for the diagnosis of PJI were 0.89 (95% confidence interval (CI), 0.83 to 0.93) and 0.96 (95% CI, 0.95 to 0.97), respectively. The value of the pooled diagnostic odds ratios (DOR) of alpha-defensin for PJI was 209.14 (95% CI, 97.31 to 449.50), and the area under the curve (AUC) was 0.98 (95% CI, 0.96 to 0.99). The pooled sensitivity and specificity of leukocyte esterase (17 studies) for the diagnosis of PJI were 0.90 (95% CI, 0.84 to 0.95) and 0.96 (95% CI, 0.93 to 0.97), respectively. The value of the DOR of leukocyte esterase for PJI was 203.23 (95% CI, 96.14 to 429.61), and the AUC was 0.98 (95% CI, 0.96 to 0.99). Based on the results of our meta-analysis, we can conclude that alpha-defensin and leukocyte esterase are valuable synovial fluid markers for identifying PJI with comparable high diagnostic accuracy.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Joints/pathology , Prosthesis-Related Infections/pathology , alpha-Defensins/metabolism , Aged , Humans , Middle Aged , Prosthesis-Related Infections/diagnosis , ROC Curve
12.
FASEB J ; 34(11): 14302-14317, 2020 11.
Article in English | MEDLINE | ID: mdl-32931052

ABSTRACT

Particles released from implants cause inflammatory bone loss, which is a key factor in aseptic loosening, the most common reason for joint replacement failure. With the anticipated increased incidence of total joint replacement in the next decade, implant failure will continue to burden patients. The gut microbiome is increasingly recognized as an important factor in bone physiology, however, its role in implant loosening is currently unknown. We tested the hypothesis that implant loosening is associated with changes in the gut microbiota in a preclinical model. When the particle challenge caused local joint inflammation, decreased peri-implant bone volume, and decreased implant fixation, the gut microbiota was affected. When the particle challenge did not cause this triad of local effects, the gut microbiota was not affected. Our results suggest that cross-talk between these compartments is a previously unrecognized mechanism of failure following total joint replacement.


Subject(s)
Gastrointestinal Microbiome , Inflammation/pathology , Osteolysis/pathology , Prostheses and Implants/adverse effects , Prosthesis-Related Infections/pathology , Animals , Inflammation/etiology , Male , Osteolysis/etiology , Prosthesis-Related Infections/etiology , Rats
13.
J Card Surg ; 35(11): 3025-3033, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32827165

ABSTRACT

BACKGROUND: Patients with prosthetic heart valves (PHV) are at an increased risk of endocarditis and dysfunction. Knowledge about the etiology of dysfunction and extent of endocarditis can have distinct treatment implications. Echocardiography has limitations due to PHV-related artifacts. We hypothesized that computed tomography (CT) will have incremental value over echocardiography for evaluation of PHV abnormalities with surgical findings as the reference standard. METHODS: Consecutive patients with PHV that had a reoperation for valve replacement, had a contrast chest CT and echocardiogram within 1 year of the reoperation, between 2010 and 2018 at a single academic center formed the study cohort. CTs and echocardiograms were assessed for potential etiologies of dysfunction (valve degeneration, pannus and thrombus); and for extent of endocarditis (vegetation, abscess, and pseudoaneurysm). RESULTS: Seventy-three patients (65.8% male, mean age 62.1 ± 16.5 years) formed the study cohort. The indication for reoperation was PHV dysfunction in 51 and PHV endocarditis in 22. Compared to echocardiography, CT diagnosed the etiology of PHV dysfunction in 17 (33.3%) more patients (9 valve degeneration, 8 pannus). In the PHV endocarditis cohort, CT failed to detect one vegetation and one abscess, whereas echocardiography failed to detect 1 abscess. In combination, CT and echocardiography demonstrated all the vegetations and abscesses. CONCLUSION: CT may provide superior characterization in comparison to echocardiography for the identification of the cause of prosthetic valve dysfunction, and complementary information to echocardiography for the evaluation of prosthetic valve endocarditis.


Subject(s)
Endocarditis/diagnostic imaging , Endocarditis/etiology , Heart Valve Prosthesis/adverse effects , Prosthesis Failure/adverse effects , Prosthesis-Related Infections/diagnostic imaging , Prosthesis-Related Infections/etiology , Tomography, X-Ray Computed , Aged , Echocardiography , Endocarditis/pathology , Endocarditis/surgery , Female , Heart Valves/diagnostic imaging , Heart Valves/pathology , Heart Valves/surgery , Humans , Male , Middle Aged , Prosthesis-Related Infections/pathology , Prosthesis-Related Infections/surgery , Reoperation , Retrospective Studies , Time Factors
14.
Methodist Debakey Cardiovasc J ; 16(2): 122-129, 2020.
Article in English | MEDLINE | ID: mdl-32670472

ABSTRACT

With its high temporal and spatial resolution and relatively low radiation exposure, positron emission tomography (PET) is increasingly being used in the management of cardiac patients, particularly those with inflammatory cardiomyopathies such as sarcoidosis. This review discusses the role of PET imaging in assessing myocardial viability, inflammatory cardiomyopathies, and endocarditis; describes the different protocols needed to acquire images for specific imaging tests; and examines imaging interpretation for each image dataset-including identification of the mismatch defect in viability imaging, which is associated with significant improvement in LV function after revascularization. We also review the role of fluorodeoxyglucose PET in cardiac sarcoidosis diagnosis, the complementary role of magnetic resonance imaging in inflammatory cardiomyopathy, and the emerging use of cardiac PET in prosthetic valve endocarditis.


Subject(s)
Cardiomyopathies/diagnostic imaging , Endocarditis/diagnostic imaging , Myocardium/pathology , Positron-Emission Tomography , Prosthesis-Related Infections/diagnostic imaging , Sarcoidosis/diagnostic imaging , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Endocarditis/pathology , Endocarditis/physiopathology , Heart Valve Prosthesis/adverse effects , Humans , Magnetic Resonance Imaging , Predictive Value of Tests , Prosthesis-Related Infections/pathology , Prosthesis-Related Infections/physiopathology , Sarcoidosis/pathology , Sarcoidosis/physiopathology , Tissue Survival , Ventricular Function, Left
15.
Bone Joint J ; 102-B(7): 899-903, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32600139

ABSTRACT

AIMS: To evaluate the histopathological examination of peri-implant tissue samples as a technique in the diagnosis of postoperative spinal implant infection (PSII). METHODS: This was a retrospective analysis. Patients who underwent revision spinal surgery at our institution were recruited for this study. PSII was diagnosed by clinical signs, histopathology, and microbiological examination of intraoperatively collected samples. Histopathology was defined as the gold standard. The sensitivity for histopathology was calculated. A total of 47 patients with PSII and at least one microbiological and histopathological sample were included in the study. RESULTS: PSII occurred in approximately 28% of the study population. Histopathology showed a sensitivity of 51.1% in the diagnosis of PSII. The most commonly found pathogens were Cutibacterium acnes and gram-positive staphylococci. CONCLUSION: Histopathology has low sensitivity for detecting PSII. In particular, infections caused by low-virulence microorganisms are insufficiently detected by histopathology. Cite this article: Bone Joint J 2020;102-B(7):899-903.


Subject(s)
Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/pathology , Spine/surgery , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Humans , Male , Middle Aged , Propionibacterium acnes/isolation & purification , Reoperation , Retrospective Studies , Sensitivity and Specificity , Staphylococcus/isolation & purification
16.
Mater Sci Eng C Mater Biol Appl ; 109: 110573, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32228937

ABSTRACT

Implant associated infections can result in devastating consequences for patients. One major cause is the formation of bacterial biofilms, which result in increased resistance against antimicrobial therapeutics. A reduction of implant associated infections can be achieved by functionalization of implant surfaces. The generation of three dimensional surface structures by femtosecond laser ablation is one method to fabricate bacterial repellent large scaled surfaces without altering the material chemical composition. The challenge is to reduce bacterial growth while improving cellular ongrowth. For this purpose, spike structures were created as small as possible by used fabrication method on cubic Ti90/Al6/V4-rods and their effectiveness against bacterial colonization was compared to unstructured Ti90/Al6/V4-rods. Rods were implanted in the rat tibia and infected intraoperatively with 103 CFU of Staphylococcus aureus. Besides clinical behaviour and lameness, the vital bacterial biomass, morphological appearance and the volume of eukaryotic cells were determined on the implant surface after 21 days. Bone alterations were examined by radiological and histological techniques. Unexpectedly, the laser-structured implants did not show a lower bacterial load on the implant surface and less severe infection related bone and tissue alterations compared to the group without structuring. Simultaneously, a better bony integration and a higher cellular colonization with eukaryotic cells was detected on the laser-structured implants. These findings don't support the previous in vitro results. Nevertheless, the strong integration into the bone is a powerful argument for further surface modifications focussing on the improvement of the antibacterial effect. Additionally, our results underline the need for in vivo testing of new materials prior to clinical use.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Implants, Experimental/microbiology , Prosthesis-Related Infections , Staphylococcal Infections , Staphylococcus aureus/physiology , Animals , Bacterial Adhesion/drug effects , Lasers , Male , Prosthesis-Related Infections/drug therapy , Prosthesis-Related Infections/metabolism , Prosthesis-Related Infections/pathology , Rats , Rats, Inbred Lew , Staphylococcal Infections/drug therapy , Staphylococcal Infections/metabolism , Staphylococcal Infections/pathology , Uracil/analogs & derivatives
17.
J Orthop Surg Res ; 15(1): 90, 2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32131862

ABSTRACT

BACKGROUND: Orthopedic metal implants are notoriously associated with release of metallic ions able to cause biological adverse reactions which might lead to implant loosening and failure. To limit any possible adverse reactions, ceramic coatings for orthopedic metal implants have been introduced. However, information regarding the interaction of these coatings with microbes responsible for periprosthetic joint infections (PJIs) is lacking. Hence, the aim of the present in vitro study is to assess the microbial affinity to a titanium-niobium nitride (TiNbN) coating. METHODS: Adhesion and biofilm formation of clinical isolates of Staphylococcus aureus, Staphylococcus epidermidis, Pseudomonas aeruginosa, and Cutibacterium acnes were assessed on TiNbN-coated titanium discs in comparison with uncoated titanium and cobalt-chrome alloys discs, with either smooth or rough surfaces. Bacterial adhesion was performed by counting adhered bacteria in the first hours of incubation, and the biofilm formation was performed by means of a spectrophotometric assay and by confocal laser scan microscopy after 72 hours of incubation. RESULTS: Overall, Staphylococcus aureus and Staphylococcus epidermidis, among the most common bacteria responsible for PJIs, displayed a significantly decreased attachment in the first hours of contact and, when cultured in presence of TiNbN coating, in comparison with CoCrMo. Biofilm formation of the four tested strains was comparable on all alloys. CONCLUSIONS: Although the onset of a PJI is more complex than in an in vitro scenario, these findings suggest that TiNbN-coated orthopedic implants do not increase PJIs risk while ameliorating tribological and surface properties could represent a valid choice to limit possible complications such as metal hypersensitivity.


Subject(s)
Alloys/administration & dosage , Bacterial Adhesion/physiology , Biocompatible Materials/administration & dosage , Biofilms/growth & development , Prosthesis-Related Infections/pathology , Staphylococcal Infections/pathology , Ceramics/therapeutic use , Humans , Microscopy, Confocal/methods , Propionibacteriaceae/growth & development , Propionibacteriaceae/isolation & purification , Prosthesis-Related Infections/prevention & control , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/isolation & purification , Staphylococcal Infections/prevention & control , Staphylococcus aureus/growth & development , Staphylococcus aureus/isolation & purification , Staphylococcus epidermidis/growth & development , Staphylococcus epidermidis/isolation & purification
18.
Mater Sci Eng C Mater Biol Appl ; 110: 110648, 2020 May.
Article in English | MEDLINE | ID: mdl-32204079

ABSTRACT

Self-assembled peptide gels have generated interest as antibacterial materials to prevent biomaterial-related infections but these peptides are often associated with poor proteolytic stability. Efforts have been made to stabilize peptides by incorporating non-natural amino acids and/or linkages but complexation with polymers have not been explored. Therefore, we developed self-assembled peptide/chitosan gels, Boc-D-Phe-γ4-L-Phe-PEA (NH007)/chitosan and Boc-L-Phe-γ4-L-Phe-PEA (NH009)/chitosan, by complexing dipeptide NH007 or NH009 with chitosan in DMSO:acetic acid. The gels were characterized using SEM, FTIR, contact angle, and rheology data and found to exhibit excellent viscoelastic and self-healing characteristics. Complexation with chitosan led to an increase in stability against proteolytic degradation. Peptide/chitosan gels showed broad spectrum antibacterial activities against Gram-negative and Gram-positive bacteria, such as Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, and Bacillus subtilis at a high inoculum of 107-108 cfu/mL. NH007/chitosan gels showed 70-75% inhibition, whereas NH009/chitosan showed 78-81% inhibition and NH009/chitosan gels, in particular, showed strong antibacterial activity against pathogenic strain of P. aeruginosa. A unique feature of these gels is that the antibacterial activities did not decrease gradually but were sustained for up to 48 h. The mechanistic studies using SEM and HR-TEM indicated interaction of gels with bacterial membrane components, leading to cell lysis. The MTT and LDH assays indicated >90% cell viability and only 8-10% toxicity towards NIH 3T3 fibroblast cells. Thus, peptide/chitosan gels developed in the present work showed improved proteolytic stability and sustained antibacterial activities and, therefore, may be used for preventing biomaterial-related infections.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Biocompatible Materials/adverse effects , Chitosan/therapeutic use , Oligopeptides/therapeutic use , Prosthesis-Related Infections/drug therapy , Animals , Anti-Bacterial Agents/pharmacology , Cell Death/drug effects , Cell Survival/drug effects , Chitosan/chemistry , Elasticity , Gels , Mice , Microbial Sensitivity Tests , NIH 3T3 Cells , Oligopeptides/chemistry , Prosthesis-Related Infections/pathology , Rheology , Viscosity
19.
Sci Rep ; 10(1): 2985, 2020 02 19.
Article in English | MEDLINE | ID: mdl-32076089

ABSTRACT

Identification of bacteria causing tissue infections can be comprehensive and, in the cases of non- or slow-growing bacteria, near impossible with conventional methods. Performing shotgun metagenomic sequencing on bacterial DNA extracted directly from the infected tissue may improve time to diagnosis and targeted treatment considerably. However, infected tissue consists mainly of human DNA (hDNA) which hampers bacterial identification. In this proof of concept study, we present a modified version of the Ultra-Deep Microbiome Prep kit for DNA extraction procedure, removing additional human DNA. Tissue biopsies from 3 patients with orthopedic implant-related infections containing varying degrees of Staphylococcus aureus were included. Subsequent DNA shotgun metagenomic sequencing using Oxford Nanopore Technologies' (ONT) MinION platform and ONTs EPI2ME WIMP and ARMA bioinformatic workflows for microbe and antibiotic resistance genes identification, respectively. The modified DNA extraction protocol led to an additional ~10-fold reduction of human DNA while preserving S. aureus DNA. Including the DNA sequencing and bioinformatics analyses, the presented protocol has the potential of identifying the infection-causing pathogen in infected tissue within 7 hours after biopsy. However, due to low number of S. aureus reads, positive identification of antibiotic resistance genes was not possible.


Subject(s)
DNA, Bacterial/isolation & purification , Metagenomics/instrumentation , Reagent Kits, Diagnostic , Staphylococcal Infections/diagnosis , Staphylococcus aureus/isolation & purification , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biopsy , Drug Resistance, Bacterial/genetics , High-Throughput Nucleotide Sequencing , Humans , Metagenome/genetics , Nanopore Sequencing , Proof of Concept Study , Prosthesis-Related Infections/diagnosis , Prosthesis-Related Infections/drug therapy , Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/pathology , Sequence Analysis, DNA , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics
20.
Sci Rep ; 10(1): 2690, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32060394

ABSTRACT

This retrospective study aimed to clarify the short- and mid-term outcomes of elderly patients who underwent surgery to treat left-sided native valve infective endocarditis (LSNIE). Between July 2005 and September 2015, 179 patients underwent surgical treatment for active LSNIE at a single institution. Patients were classified into two groups: ≥65 years (elderly group) and <65 years (non-elderly group). Clinical features, surgical information, postoperative complications, and three-year survival rates were compared. The average ages were 74.2 ± 6.4 and 45.2 ± 12.6 years in the elderly and non-elderly groups, respectively. The elderly group had a higher predicted mortality rate and a lower incidence of preoperative septic emboli-related complications. Echocardiographic assessments of infected valves were generally homogenous between the groups. The elderly patients had a higher in-hospital mortality rate than the non-elderly patients (26.3% vs. 5.7%, P = 0.001). For patients who survived to discharge, the three-year cumulative survival rates were 75.0% ± 8.2% and 81.2% ± 3.4% in the elderly and non-elderly groups, respectively (P = 0.484). In conclusion, elderly patients are at a higher risk of in-hospital mortality after surgery for LSNIE. However, once elderly patients are stabilized by surgical treatment and survive to discharge, the mid-term outcomes are promising.


Subject(s)
Endocarditis, Bacterial/surgery , Heart Valve Prosthesis Implantation/adverse effects , Prosthesis-Related Infections/surgery , Staphylococcal Infections/surgery , Adult , Aged , Aged, 80 and over , Echocardiography , Endocarditis, Bacterial/complications , Endocarditis, Bacterial/microbiology , Endocarditis, Bacterial/pathology , Female , Heart Valve Prosthesis/microbiology , Heart Valve Prosthesis Implantation/methods , Hospital Mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prosthesis-Related Infections/complications , Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/pathology , Retrospective Studies , Risk Factors , Staphylococcal Infections/complications , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...