SwePub
Sök i SwePub databas

  Extended search

Träfflista för sökning "WFRF:(Jin Tao 1973) srt2:(2015-2019)"

Search: WFRF:(Jin Tao 1973) > (2015-2019)

  • Result 1-10 of 20
Sort/group result
   
EnumerationReferenceCoverFind
1.
  • Ali, Abukar, 1988, et al. (author)
  • Antibiotic-killed Staphylococcus aureus induces destructive arthritis in mice.
  • 2015
  • In: Arthritis & rheumatology (Hoboken, N.J.). - : Wiley. - 2326-5205 .- 2326-5191. ; 67:1, s. 107-116
  • Journal article (peer-reviewed)abstract
    • Objective: Permanent reduction in joint function is a severe post-infectious complication in patients with Staphylococcus aureus septic arthritis. This reduction in joint function might be caused by persistent joint inflammation after the adequate eradication of bacteria by antibiotics. Methods: We studied whether antibiotic-killed S. aureus induced joint inflammation in mice and elucidated the molecular and cellular mechanism of this type of arthritis. Results: The intraarticular injection of antibiotic-killed S. aureus induced mild to moderate synovitis and bone erosions that lasted for a minimum of 14 days. The frequency and severity of synovitis were significantly reduced in tumor necrosis factor receptor 1 (TNFR1), receptor for Advanced Glycation End Products (RAGE), and toll like receptor 2 (TLR2) knockout mice compared with wild-type animals. The combined depletion of monocytes and neutrophils resulted in a significantly lower frequency of synovitis. Among bacterial factors, insoluble cell debris played a more important role than bacterial DNA or soluble components in inducing joint inflammation. Importantly, anti-TNF therapy abrogated the joint inflammation induced by antibiotic-killed S. aureus. Conclusion: Antibiotic-killed S. aureus induced and maintained the joint inflammation that is mediated through TLR2, TNFR1, and RAGE receptor. The cross-talk between neutrophils and monocytes is responsible for this type of arthritis. Anti-TNF therapy might be used as a novel therapeutic strategy, in combination with antibiotics, to treat staphylococcal septic arthritis. © 2014 American College of Rheumatology.
  •  
2.
  • Ali, Abukar, 1988, et al. (author)
  • CTLA4-Ig but not anti-TNF therapy promotes staphylococcal septic arthritis in mice.
  • 2015
  • In: The Journal of infectious diseases. - : Oxford University Press (OUP). - 1537-6613 .- 0022-1899. ; 212:8, s. 1308-1316
  • Journal article (peer-reviewed)abstract
    • The development of biologics has greatly increased the quality of life as well as the life expectancy of many RA patients. However, a large number of these patients are at an increased risk of developing serious infections. The aim of this study was to examine differential effects of anti-TNF versus CTLA4-Ig treatment on both immunological response and host defense in a murine model of septic arthritis.
  •  
3.
  • Ali, Abukar, 1988, et al. (author)
  • IL-1 Receptor Antagonist Treatment Aggravates Staphylococcal Septic Arthritis and Sepsis in Mice.
  • 2015
  • In: PloS one. - : Public Library of Science (PLoS). - 1932-6203. ; 10:7
  • Journal article (peer-reviewed)abstract
    • Interleukin-1 receptor antagonist (IL-1Ra) is the primary therapy against autoinflammatory syndromes with robust efficacy in reducing systemic inflammation and associated organ injury. However, patients receiving IL-1Ra might be at increased risk of acquiring serious infections.
  •  
4.
  • Baranwal, Gaurav, et al. (author)
  • Impact of cell wall peptidoglycan O-acetylation on the pathogenesis of Staphylococcus aureus in septic arthritis.
  • 2017
  • In: International journal of medical microbiology : IJMM. - : Elsevier BV. - 1618-0607 .- 1438-4221. ; 307:7, s. 388-397
  • Journal article (peer-reviewed)abstract
    • Staphylococcus aureus (S. aureus) is one of the most common pathogen causing septic arthritis. To colonize the joints and establish septic arthritis this bacterium needs to resist the host innate immune responses. Lysozyme secreted by neutrophils and macrophages is an important defense protein present in the joint synovial fluids. S. aureus is known to be resistant to lysozyme due to its peptidoglycan modification by O-acetylation of N-acetyl muramic acid. In this study we have investigated the role of O-acetylated peptidoglycan in septic arthritis. Using mouse models for both local and hematogenous S. aureus arthritis we compared the onset and progress of the disease induced by O-acetyl transferase mutant and the parenteral wild type SA113 strain. The disease progression was assessed by observing the clinical parameters including body weight, arthritis, and functionality of the affected limbs. Further X-ray and histopathological examinations were performed to monitor the synovitis and bone damage. In local S. aureus arthritis model, mice inoculated with the ΔoatA strain developed milder disease (in terms of knee swelling, motor and movement functionality) compared to mice inoculated with the wild type SA113 strain. X-ray and histopathological data revealed that ΔoatA infected mice knee joints had significantly lesser joint destruction, which was accompanied by reduced bacterial load in knee joints. Similarly, in hematogenous S. aureus arthritis model, ΔoatA mutant strain induced significantly less severe clinical septic arthritis compared to its parental strain, which is in accordance with radiological findings. Our data indicate that peptidoglycan O-acetylation plays an important role in S. aureus mediated septic arthritis.
  •  
5.
  • Boldock, Emma, et al. (author)
  • Human skin commensals augment Staphylococcus aureus pathogenesis.
  • 2018
  • In: Nature microbiology. - : Springer Science and Business Media LLC. - 2058-5276. ; 3:8, s. 881-90
  • Journal article (peer-reviewed)abstract
    • All bacterial infections occur within a polymicrobial environment, from which a pathogen population emerges to establish disease within a host. Emphasis has been placed on prevention of pathogen dominance by competing microflora acting as probiotics1. Here we show that the virulence of the human pathogen Staphylococcus aureus is augmented by native, polymicrobial, commensal skin flora and individual species acting as 'proinfectious agents'. The outcome is pathogen proliferation, but not commensal. Pathogenesis augmentation can be mediated by particulate cell wall peptidoglycan, reducing the S. aureus infectious dose by over 1,000-fold. This phenomenon occurs using a range of S. aureus strains and infection models and is not mediated by established receptor-mediated pathways including Nod1, Nod2, Myd88 and the NLPR3 inflammasome. During mouse sepsis, augmentation depends on liver-resident macrophages (Kupffer cells) that capture and internalize both the pathogen and the proinfectious agent, leading to reduced production of reactive oxygen species, pathogen survival and subsequent multiple liver abscess formation. The augmented infection model more closely resembles the natural situation and establishes the role of resident environmental microflora in the initiation of disease by an invading pathogen. As the human microflora is ubiquitous2, its role in increasing susceptibility to infection by S. aureus highlights potential strategies for disease prevention.
  •  
6.
  • Elmwall, Jonas, et al. (author)
  • Galectin-3 Is a Target for Proteases Involved in the Virulence of Staphylococcus aureus
  • 2017
  • In: Infection and Immunity. - : American Society for Microbiology. - 0019-9567 .- 1098-5522. ; 85:7
  • Journal article (peer-reviewed)abstract
    • Staphylococcus aureus is a major cause of skin and soft tissue infection. The bacterium expresses four major proteases that are emerging as virulence factors: aureolysin (Aur), V8 protease (SspA), staphopain A (ScpA), and staphopain B (SspB). We hypothesized that human galectin-3, a beta-galactoside-binding lectin involved in immune regulation and antimicrobial defense, is a target for these proteases and that proteolysis of galectin-3 is a novel immune evasion mechanism. Indeed, supernatants from laboratory strains and clinical isolates of S. aureus caused galectin-3 degradation. Similar proteolytic capacities were found in Staphylococcus epidermidis isolates but not in Staphylococcus saprophyticus. Galectin-3-induced activation of the neutrophil NADPH oxidase was abrogated by bacterium-derived proteolysis of galectin-3, and SspB was identified as the major protease responsible. The impact of galectin-3 and protease expression on S. aureus virulence was studied in a murine skin infection model. In galectin-3 (+)/(+) mice, SspB-expressing S. aureus caused larger lesions and resulted in higher bacterial loads than protease-lacking bacteria. No such difference in bacterial load or lesion size was detected in galectin-3 (+)/(+) mice, which overall showed smaller lesion sizes than the galectin-3 (+)/(+) animals. In conclusion, the staphylococcal protease SspB inactivates galectin-3, abrogating its stimulation of oxygen radical production in human neutrophils and increasing tissue damage during skin infection.
  •  
7.
  • Fatima, Farah, et al. (author)
  • Radiological features of experimental staphylococcal septic arthritis by micro computed tomography scan.
  • 2017
  • In: PloS one. - : Public Library of Science (PLoS). - 1932-6203. ; 12:2
  • Journal article (peer-reviewed)abstract
    • Permanent joint dysfunction due to bone destruction occurs in up to 50% of patients with septic arthritis. Recently, imaging technologies such as micro computed tomography (μCT) scan have been widely used for preclinical models of autoimmune joint disorders. However, the radiological features of septic arthritis in mice are still largely unknown.NMRI mice were intravenously or intra-articularly inoculated with S. aureus Newman or LS-1 strain. The radiological and clinical signs of septic arthritis were followed for 10 days using μCT. We assessed the correlations between joint radiological changes and clinical signs, histological changes, and serum levels of cytokines.On days 5-7 after intravenous infection, bone destruction verified by μCT became evident in most of the infected joints. Radiological signs of bone destruction were dependent on the bacterial dose. The site most commonly affected by septic arthritis was the distal femur in knees. The bone destruction detected by μCT was positively correlated with histological changes in both local and hematogenous septic arthritis. The serum levels of IL-6 were significantly correlated with the severity of joint destruction.μCT is a sensitive method for monitoring disease progression and determining the severity of bone destruction in a mouse model of septic arthritis. IL-6 may be used as a biomarker for bone destruction in septic arthritis.
  •  
8.
  • Jin, Tao, 1973, et al. (author)
  • A novel mouse model for septic arthritis induced by Pseudomonas aeruginosa
  • 2019
  • In: Scientific Reports. - : Springer Science and Business Media LLC. - 2045-2322. ; 9
  • Journal article (peer-reviewed)abstract
    • Septic arthritis is one of the most aggressive joint diseases. Although caused predominantly by S. aureus, Gram-negative bacteria, Pseudomonas aeruginosa among them, account for a significant percentage of the causal agents of septic arthritis. However, septic arthritis caused by P. aeruginosa has not been studied thus far, due to lack of an animal model. NMRI mice were inoculated with different doses of P. aeruginosa. The clinical course of septic arthritis and radiological changes of joints were examined. Furthermore, the host molecular and cellular mechanisms involved in P. aeruginosa-induced septic arthritis were investigated. Inoculation of mice with P. aeruginosa caused septic arthritis in a dose-dependent manner. Neutrophil depletion led to higher mortality and more severe joint destruction (p < 0.01). In contrast, monocyte depletion resulted in higher mortality (p < 0.05) but similar arthritis severity compared to controls. Mice depleted of CD4+ T-cells inoculated with P. aeruginosa displayed less severe bone damage (p < 0.05). For the first time, a mouse model for P. aeruginosa septic arthritis is presented. Our data demonstrate that neutrophils play a protective role in P. aeruginosa septic arthritis. Monocytes/macrophages, on the other hand, are only essential in preventing P. aeruginosa-induced mortality. Finally, CD4+ T-cells are pathogenic in P. aeruginosa septic arthritis.
  •  
9.
  •  
10.
  • Kwiecinski, Jakub, 1985, et al. (author)
  • Biofilm formation by Staphylococcus aureus clinical isolates correlates with the infection type.
  • 2019
  • In: Infectious diseases. - : Informa UK Limited. - 2374-4243 .- 2374-4235. ; 51:6, s. 446-451
  • Journal article (peer-reviewed)abstract
    • Biofilms are involved in many Staphylococcus aureus infections, but relation of biofilm formation and the infection types or the clinical outcomes remain unclear.We measured biofilm formation, with a microtiter plate assay, of a collection of methicillin-sensitive clinical isolates from 159 invasive S. aureus infections, encompassing all cases occurring within a hospital catchment area during two years, and from additional 49 non-invasive skin infections from the same region. Results were related to available clinical and microbiological documentation.Isolates from medical device infections (intravenous line-associated and prosthetic joint infections), as well as isolates from superficial skin infections, were particularly proficient in forming biofilms. No increased biofilm-forming capacity was seen in isolates from endocarditis, osteomyelitis, or other infections. There was also a correlation of biofilm formation with the agr type of the isolates. Thicker biofilms were more resistant to antibiotic treatment in vitro. No correlation between biofilm formation and clinical outcomes was noted.S. aureus isolates from 'classical' biofilm-related infections, but also from superficial skin infections, are especially proficient in forming biofilms. There is, however, no obvious relation of biofilm-forming capacity of isolates and the clinical outcome of the infection, and more studies on this issue are needed.
  •  
Skapa referenser, mejla, bekava och länka
  • Result 1-10 of 20
Type of publication
journal article (20)
Type of content
peer-reviewed (20)
Author/Editor
Jin, Tao, 1973 (20)
Na, Manli (13)
Ali, Abukar, 1988 (11)
Mohammad, Majd (9)
Pullerits, Rille, 19 ... (8)
Kwiecinski, Jakub, 1 ... (7)
show more...
Josefsson, Elisabet, ... (7)
Welin, Amanda, 1983 (5)
Jarneborn, Anders (5)
Magnusson, Malin (4)
Bylund, Johan, 1975 (3)
Svensson, Mattias, 1 ... (2)
Nawaz, Muhammad (2)
Zhu, Xuefeng (2)
Schwarze, Jan-Christ ... (2)
Karlsson, Anna, 1967 (2)
Götz, Friedrich (2)
Valadi, Hadi, 1963 (2)
Sunnerhagen, Per, 19 ... (1)
Moore, Edward R.B. 1 ... (1)
Bokarewa, Maria, 196 ... (1)
Svensson, Camilla (1)
Catrina, Anca I (1)
Klareskog, Lars (1)
McCoy, Kathy D (1)
Verhamme, Peter (1)
Lycke, Nils Y, 1954 (1)
Fei, Y (1)
Svensson, Mattias N ... (1)
Gjertsson, Inger, 19 ... (1)
Hölttä, Mikko (1)
Erlandsson, Malin, 1 ... (1)
Lindholm, Catharina, ... (1)
Wang, Xiaoyang, 1965 (1)
Johansson, Bengt R, ... (1)
Iwakura, Yoichiro (1)
Mårtensson, Inga-Lil ... (1)
Jacobsson, Gunnar, 1 ... (1)
Nguyen, Minh-Thu (1)
Malmström, Vivianne (1)
Johansson, Svante (1)
Engdahl, Cecilia, 19 ... (1)
Pekna, Marcela, 1966 (1)
Stokowska, Anna (1)
Kahlmeter, Gunnar (1)
Sävman, Karin, 1960 (1)
Forsman, Huamei (1)
Angerfors, Annelie (1)
Camponeschi, Alessan ... (1)
Horswill, Alexander ... (1)
show less...
University
University of Gothenburg (20)
Umeå University (2)
Karolinska Institutet (1)
Language
English (20)
Research subject (UKÄ/SCB)
Medical and Health Sciences (20)

Year

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Close

Copy and save the link in order to return to this view