Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
Add more filters

Publication year range
1.
BJU Int ; 134(1): 72-80, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38459675

ABSTRACT

OBJECTIVES: To investigate the ability of propolis-coated ureteric stents to solve complications, especially urinary tract infections (UTIs) and crusting, in patients with long-term indwelling ureteric stents through antimicrobial and anti-calculus activities. MATERIALS AND METHODS: Polyurethane (PU) ureteric stents were immersed in the ethanol extract of propolis (EEP), a well-known antimicrobial honeybee product, and subjected to chemical, hydrophilic, and seismic tests. The antimicrobial activity of the EEP coating was then examined by in vitro investigation. Proteus mirabilis infection was induced in rats within uncoated and EEP-coated groups, and the infection, stone formation, and inflammation were monitored at various time points. RESULTS: The characterisation results showed that the hydrophilicity and stability of the EEP surface improved. In vitro tests revealed that the EEP coating was biocompatible, could eliminate >90% of bacteria biofilms attached to the stent and could maintain bacteriostatic properties for up to 3 months. The in vivo experiment revealed that the EEP-coating significantly reduced the amount of bacteria, stones, and salt deposits on the surface of the ureteric stents and decreased inflammation in the host tissue. CONCLUSIONS: Compared with clinically used PU stents, EEP-coated ureteric stents could better mitigate infections and prevent encrustation. Thus, this study demonstrated that propolis is a promising natural dressing material for ureteric stents.


Subject(s)
Anti-Bacterial Agents , Coated Materials, Biocompatible , Propolis , Stents , Ureter , Animals , Rats , Propolis/pharmacology , Anti-Bacterial Agents/pharmacology , Coated Materials, Biocompatible/pharmacology , Proteus mirabilis/drug effects , Male , Urinary Tract Infections/prevention & control , Rats, Sprague-Dawley , Biofilms/drug effects , Proteus Infections/prevention & control , Polyurethanes
2.
J Appl Microbiol ; 134(6)2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37296327

ABSTRACT

AIMS: The care of patients undergoing long-term urethral catheterization is frequently complicated by Proteus mirabilis infection. This organism forms dense, crystalline biofilms, which block catheters leading to serious clinical conditions. However, there are currently no truly effective approaches to control this problem. Here, we describe the development of a novel theranostic catheter coating, to simultaneously provide early warning of blockage, and actively delay crystalline biofilm formation. METHODS AND RESULTS: The coating comprises of a pH sensitive upper polymer layer (poly(methyl methacrylate-co-methacrylic acid); Eudragit S 100®) and a hydrogel base layer of poly(vinyl alcohol), which is loaded with therapeutic agents (acetohydroxamic acid or ciprofloxacin hydrochloride) and a fluorescent dye, 5(6)-carboxyfluorescein (CF). The elevation of urinary pH due to P. mirabilis urease activity results in the dissolution of the upper layer and release of cargo agents contained in the base layer. Experiments using in vitro models, which were representative of P. mirabilis catheter-associated urinary tract infections, demonstrated that these coatings significantly delay time taken for catheters to block. Coatings containing both CF dye and ciprofloxacin HCl were able to provide an average of ca. 79 h advanced warning of blockage and extend catheter lifespan ca. 3.40-fold. CONCLUSIONS: This study has demonstrated the potential for theranostic, infection-responsive coatings to form a promising approach to combat catheter encrustation and actively delay blockage.


Subject(s)
Proteus Infections , Urinary Tract Infections , Humans , Urinary Catheters , Urinary Catheterization/adverse effects , Proteus Infections/prevention & control , Proteus Infections/etiology , Proteus mirabilis , Urinary Tract Infections/prevention & control , Biofilms
3.
Microb Pathog ; 132: 293-301, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31082531

ABSTRACT

Stone formation and catheter blockage are major complications of Proteus UTIs. In this study, we investigated the ability of allicin to inhibit P. mirabilis-induced struvite crystallization and catheter blockage using a synthetic bladder model. Struvite crystallization inhibition study was carried out using P. mirabilis lysate as urease enzyme source in synthetic urine (SU). Struvite productions were monitored by phase contrast light microscopy and measurements of pH, Mg2+ and Ca2+ precipitation and turbidity. A catheter blockage study was performed in a synthetic bladder model mimicking natural UTI in the presence of allicin at sub-MIC concentrations (MIC = 64 µg/ml). The results of crystallization study showed that allicin inhibited pH rise and consequently turbidity and precipitation of ions in a dose-dependent manner. The results of catheter blockage study showed that allicin at sub-MIC concentrations (2, 4, 8 µg/ml) significantly increased the time for catheter blockage to occur to 61, 74 and 92 h respectively compared to allicin-free control (48 h). In a similar way, the results showed that allicin delayed the increase of SU pH level in bladder model in a dose-dependent manner compared to allicin-free control. The results also showed that following the increase of allicin concentration, Mg2+ and Ca2+ deposition in catheters were much lower compared to allicin-free control, further confirmed by direct observation of the catheters' eyehole and cross sections. We conclude that allicin prevents the formation of Proteus-induced urinary crystals and the blockage of catheters by delaying pH increase and lowering Mg2+ and Ca2+ deposition in a dose-dependent manner.


Subject(s)
Proteus Infections/prevention & control , Proteus/drug effects , Sulfinic Acids/pharmacology , Urinary Bladder/microbiology , Calcium/metabolism , Crystallization , Disulfides , Dose-Response Relationship, Drug , Humans , Hydrogen-Ion Concentration , Magnesium/metabolism , Microbial Sensitivity Tests , Proteus/growth & development , Proteus mirabilis/drug effects , Proteus mirabilis/growth & development , Urease , Urinary Tract Infections/microbiology , Urinary Tract Infections/prevention & control , Urine
4.
Postepy Hig Med Dosw (Online) ; 70(0): 1032-1043, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-27708208

ABSTRACT

Urinary tract infections (UTIs) pose a threat especially to women, the individuals with weakened immunity or with abnormalities in the urinary tract as well as to hospitalized and catheterized patients. The bacteria from the genus Proteus, especially P. mirabilis, are important UTI pathogenic factors. They frequently cause chronic, recurrent or severely complicated infections, resulting in the urinary stones production due to urease and other virulence factors. The ability to survive inside the stones and the increasing antibiotic resistance make it difficult to eradicate the bacteria from the urinary tract. A good solution to the problem may be the vaccination which obtained the interest from the surveyed persons, in spite of the antivaccination attitudes visible also in Poland. Currently, there are four vaccines available, composed of killed cells of different uropathogens, including Proteus spp. They are administrated intranassaly or vaginally and require many booster doses. They decrease the probability of reinfection in patients suffering from recurrent UTIs but the mechanisms of the immune response have not been exactly defined. Promising results were obtained in the studies on a mice model concerning the subunit, conjugated vaccines in which various P. mirabilis surface antigens (with the exception of flagellin) were successfully employed. Hitherto, the best results were obtained by the intranasal vaccinations, using MR/P fimbriae antigens with MPL or cholera toxin adjuvants and the antigens expressed in Lactococcus lactis or Salmonella Typhimurium. It led to an increase in the levels of the specific serum and mucosal antibodies resulting in the protection against P. mirabilis UTIs.


Subject(s)
Bacterial Vaccines/therapeutic use , Proteus Infections/prevention & control , Proteus mirabilis , Urinary Tract Infections/prevention & control , Adjuvants, Immunologic , Female , Humans , Male , Poland , Proteus Infections/immunology , Urinary Tract Infections/microbiology
5.
Protein Expr Purif ; 105: 33-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25317910

ABSTRACT

Proteus mirabilis (P. mirabilis) is a zoonotic pathogen that has recently presented a rising infection rate in the poultry industry. To develop an effective vaccine to protect chickens against P. mirabilis infection, OmpA, one of the major outer membrane proteins of P. mirabilis, was expressed in Pichia pastoris. The concentration of the expressed recombinant OmpA protein reached 8.0µg/mL after induction for 96h with 1.0% methanol in the culture. In addition, OmpA protein was confirmed by SDS-PAGE and Western blot analysis using the antibody against Escherichia coli-expressed OmpA protein. Taishan Pinus massoniana pollen polysaccharide, a known plant-derived adjuvant, was mixed into the recombinant OmpA protein to prepare the OmpA subunit vaccine. We then subcutaneously inoculated this vaccine into chickens to examine the immunoprotective effects. ELISA analysis indicated that an excellent antibody response against OmpA was elicited in the vaccinated chickens. Moreover, a high protection rate of 80.0% was observed in the vaccinated group, which was subsequently challenged with P. mirabilis. The results suggest that the eukaryotic P. mirabilis OmpA was an ideal candidate protein for developing an effective subunit vaccine against P. mirabilis infection.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Recombinant Proteins/immunology , Vaccines, Subunit/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/isolation & purification , Bacterial Outer Membrane Proteins/metabolism , Bacterial Vaccines/genetics , Bacterial Vaccines/metabolism , Chickens , Pichia/genetics , Pichia/metabolism , Proteus Infections/immunology , Proteus Infections/microbiology , Proteus Infections/prevention & control , Proteus mirabilis/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Vaccines, Subunit/genetics , Vaccines, Subunit/metabolism
6.
J Intern Med ; 276(2): 120-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24635559

ABSTRACT

This review is largely based on a previous paper published in the journal Spinal Cord. The care of many patients undergoing long-term bladder catheterization is complicated by encrustation and blockage of their Foley catheters. This problem stems from infection by urease-producing bacteria, particularly Proteus mirabilis. These organisms colonize the catheter forming an extensive biofilm; they also generate ammonia from urea, thus elevating the pH of urine. As the pH rises, crystals of calcium and magnesium phosphates precipitate in the urine and in the catheter biofilm. The continued development of this crystalline biofilm blocks the flow of urine through the catheter. Urine then either leaks along the outside of the catheter and the patient becomes incontinent or is retained causing painful distension of the bladder and reflux of urine to the kidneys. The process of crystal deposition can also initiate stone formation. Most patients suffering from recurrent catheter encrustation develop bladder stones. P. mirabilis establishes stable residence in these stones and is extremely difficult to eliminate from the catheterized urinary tract by antibiotic therapy. If blocked catheters are not identified and changed, serious symptomatic episodes of pyelonephritis, septicaemia and endotoxic shock can result. All types of Foley catheters including silver- or nitrofurazone-coated devices are vulnerable to this problem. In this review, the ways in which biofilm formation on Foley catheters is initiated by P. mirabilis will be described. The implications of understanding these mechanisms for the development of an encrustation-resistant catheter will be discussed. Finally, the way forward for the prevention and control of this problem will be considered.


Subject(s)
Biofilms/growth & development , Catheter-Related Infections/microbiology , Proteus mirabilis/pathogenicity , Urinary Catheterization/adverse effects , Urinary Catheters/adverse effects , Urinary Tract Infections/microbiology , Calcium Compounds/metabolism , Catheter-Related Infections/prevention & control , Citrates/urine , Crystallization , Humans , Magnesium Compounds/metabolism , Proteus Infections/etiology , Proteus Infections/prevention & control , Time Factors , Urinary Catheterization/instrumentation , Urinary Catheterization/methods , Urinary Tract Infections/enzymology , Urinary Tract Infections/prevention & control , Urine/chemistry , Urine/microbiology
7.
Antonie Van Leeuwenhoek ; 105(6): 1139-48, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24771125

ABSTRACT

Proteus mirabilis expresses several virulence factors including MR/P fimbriae and flagella. Bacterial flagellin has frequently shown interesting adjuvant and protective properties in vaccine formulations. However, native P. mirabilis flagellin has not been analyzed so far. Native P. mirabilis flagellin was evaluated as a protective antigen and as an adjuvant in co-immunizations with MrpA (structural subunit of MR/P fimbriae) using an ascending UTI model in the mouse. Four groups of mice were intranasally treated with either MrpA, native flagellin, both proteins and PBS. Urine and blood samples were collected before and after immunization for specific antibodies determination. Cytokine production was assessed in immunized mice splenocytes cultures. Mice were challenged with P. mirabilis, and bacteria quantified in kidneys and bladders. MrpA immunization induced serum and urine specific anti-MrpA antibodies while MrpA coadministered with native flagellin did not. None of the animals developed significant anti-flagellin antibodies. Only MrpA-immunized mice showed a significant decrease of P. mirabilis in bladders and kidneys. Instead, infection levels in MrpA-flagellin or flagellin-treated mice showed no significant differences with the control group. IL-10 was significantly induced in splenocytes of mice that received native flagellin or MrpA-flagellin. Native P. mirabilis flagellin did not protect mice against an ascending UTI. Moreover, it showed an immunomodulatory effect, neutralizing the protective role of MrpA. P. mirabilis flagellin exhibits particular immunological properties compared to other bacterial flagellins.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Flagellin/immunology , Proteus Infections/prevention & control , Proteus mirabilis/immunology , Urinary Tract Infections/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/urine , Antigens, Bacterial/administration & dosage , Bacterial Proteins/administration & dosage , Bacterial Vaccines/administration & dosage , Disease Models, Animal , Drug Antagonism , Flagellin/administration & dosage , Interleukin-10/biosynthesis , Kidney/microbiology , Leukocytes, Mononuclear/immunology , Mice , Proteus Infections/immunology , Proteus mirabilis/growth & development , Urinary Bladder/microbiology , Urinary Tract Infections/immunology
8.
Front Cell Infect Microbiol ; 13: 1216798, 2023.
Article in English | MEDLINE | ID: mdl-37965267

ABSTRACT

Introduction: Proteus mirabilis is a key pathobiont in catheter-associated urinary tract infections (CA-UTIs), which is well known to form crystalline biofilms that occlude catheters. Urease activity alkylates urine through the release of ammonia, consequentially resulting in higher levels of Mg2+ and Ca2+ and formation of crystals. In this study, we showed that N-acetyl cysteine (NAC), a thiol antioxidant, is a potent urease inhibitor that prevents crystalline biofilm formation. Methods: To quantify urease activity, Berthelot's method was done on bacterial extracts treated with NAC. We also used an in vitro catheterised glass bladder model to study the effect of NAC treatment on catheter occlusion and biofilm encrustation in P. mirabilis infections. Inductively-coupled plasma mass spectrometry (ICP-MS) was performed on catheter samples to decipher elemental profiles. Results: NAC inhibits urease activity of clinical P. mirabilis isolates at concentrations as low as 1 mM, independent of bacterial killing. The study also showed that NAC is bacteriostatic on P. mirabilis, and inhibited biofilm formation and catheter occlusion in an in vitro. A significant 4-8log10 reduction in viable bacteria was observed in catheters infected in this model. Additionally, biofilms in NAC treated catheters displayed a depletion of calcium, magnesium, or phosphates (>10 fold reduction), thus confirming the absence of any urease activity in the presence of NAC. Interestingly, we also showed that not only is NAC anti-inflammatory in bladder epithelial cells (BECs), but that it mutes its inflammatory response to urease and P. mirabilis infection by reducing the production of IL-6, IL-8 and IL-1b. Discussion: Using biochemical, microbiological and immunological techniques, this study displays the functionality of NAC in preventing catheter occlusion by inhibiting urease activity. The study also highlights NAC as a strong anti-inflammatory antibiofilm agent that can target both bacterial and host factors in the treatment of CA-UTIs.


Subject(s)
Proteus Infections , Urinary Tract Infections , Humans , Urinary Catheterization , Acetylcysteine/pharmacology , Urease , Proteus Infections/drug therapy , Proteus Infections/prevention & control , Proteus Infections/microbiology , Proteus mirabilis , Urinary Tract Infections/prevention & control , Urinary Tract Infections/microbiology , Catheters , Inflammation/prevention & control , Anti-Inflammatory Agents/pharmacology , Biofilms
10.
Br J Community Nurs ; 16(11): 528, 530-3, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22145192

ABSTRACT

Urinary tract infectious caused by Proteus are uncommon, and account for only 1-2% of community-acquired infectious. However, it is important that staff are able to recognise the features of a Proteus infection. Proteus infections, if unrecognised, can become established and cause major health problems. Proteus is more common in people who have or have had a urinary catheter. Minimizing the incidence and duration of urinary catheterization is an important part of preventing infection. Community staff who are alert to the possibility of this infection can make a huge difference to their patient's quality of life.


Subject(s)
Catheter-Related Infections/prevention & control , Proteus Infections/prevention & control , Proteus mirabilis , Urinary Catheterization/nursing , Adult , Aged , Algorithms , Catheter-Related Infections/diagnosis , Catheters, Indwelling/adverse effects , Female , Humans , Male , Proteus Infections/diagnosis , Quality of Life , Urinary Catheterization/adverse effects
11.
Infect Genet Evol ; 95: 105045, 2021 11.
Article in English | MEDLINE | ID: mdl-34428568

ABSTRACT

Multi-drug resistance of Proteus mirabilis, a frequent cause of catheter-associated urinary tract infections, renders ineffective treatment. Therefore, new advanced strategies are needed to overcome it. In the meantime, vaccination may be the most effective and promising method. In this study antigenicity, allergenicity, subcellular localization, human homology, B-cell epitopes and MHC-II binding sites, conserved domains and protein-protein interactions were predicted using different reverse vaccinology methods and bioinformatics databases to find new putative immunogenic targets against P. mirabilis. Finally, 5 putative immunogenic targets against P. mirabilis were identified. Considering all criteria, QKQ94350.1 (Cell envelope opacity-associated protein A), QKQ94681.1 (Porin), QKQ95001.1 (TonB-dependent hemoglobin/ transferrin/ lactoferrin family receptor), QKQ95221.1 (AsmA) and QKQ94335.1 (N-acetylmuramoyl-L-alanine amidase) are excellent putative immunogenic targets. Finally, a multi-epitope vaccine was designed using the conserved linear epitopes of two OMPs (QKQ94681.1 and QKQ95001.1) and N-acetylmuramoyl-L-alanine amidase (QKQ94335.1), which have promising properties for immunization. These findings can simplify the development of efficient vaccines against P. mirabilis.


Subject(s)
Bacterial Vaccines/immunology , Epitopes, B-Lymphocyte/immunology , Proteus Infections/prevention & control , Proteus mirabilis/immunology , Urinary Tract Infections/prevention & control , Computational Biology , Proteus Infections/microbiology , Urinary Tract Infections/microbiology , Vaccinology
12.
J Infect Dev Ctries ; 14(4): 366-372, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32379713

ABSTRACT

INTRODUCTION: Approximately 5% of men and 40%-50% of women have experienced urinary tract infections (UTI), which are the most common infectious diseases and nosocomial infections in humans. Proteus mirabilis is susceptible to most antibiotics, but antibiotic treatment usually causes side effects. In this research, lactic acid bacteria (LAB) was assessed for its inhibitory activity against a urinary tract pathogen. METHODOLOGY: We studied the effect of pH adjustment, heat, and enzyme treatments on the inhibitory activity of LAB strains and their supernatants, using well-diffusion and co-culture assays. In the cell culture assay, anti-adhesion and anti-invasion activities against P. mirabilis were tested with SV-HUC-1 urothelial cells. RESULTS: LAB were able to adhere to the urothelial cells and inhibited P. mirabilis growth. LAB were also able to inhibit P. mirabilis adhesion to or invasion of SV-HUC-1 urothelial cells. Finally, in the competition assay, LAB showed inhibitory effects against P. mirabilis. LAB could also inhibit the invasion of P. mirabilis into urothelial cells. CONCLUSIONS: Two LAB strains (PM206 and 229) exhibited antagonistic activity against P. mirabilis adhesion or invasion of urothelial cells in culture. In the future, probiotics may be used in food or urinary tract cleansing and could replace antibiotic treatments.


Subject(s)
Lactobacillales/physiology , Probiotics/pharmacology , Proteus Infections/prevention & control , Urinary Tract Infections/prevention & control , Urothelium/microbiology , Antibiosis , Bacterial Adhesion , Cell Line , Culture Media , Female , Humans , Proteus mirabilis/growth & development , Urinary Tract Infections/microbiology , Urothelium/cytology
13.
Infect Immun ; 77(2): 632-41, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19029299

ABSTRACT

Complicated urinary tract infections (UTI) caused by Proteus mirabilis are associated with severe pathology in the bladder and kidney. To investigate the roles of two established cytotoxins, the HpmA hemolysin, a secreted cytotoxin, and proteus toxic agglutinin (Pta), a surface-associated cytotoxin, mutant analysis was used in conjunction with a mouse model of ascending UTI. Inactivation of pta, but not inactivation of hpmA, resulted in significant decreases in the bacterial loads of the mutant in kidneys (P < 0.01) and spleens (P < 0.05) compared to the bacterial loads of the wild type; the 50% infective dose (ID(50)) of an isogenic pta mutant or hpmA pta double mutant was 100-fold higher (5 x 10(8) CFU) than the ID(50) of parent strain HI4320 (5 x 10(6) CFU). Colonization by the parent strain caused severe cystitis and interstitial nephritis as determined by histopathological examination. Mice infected with the same bacterial load of the hpmA pta double mutant showed significantly reduced pathology (P < 0.01), suggesting that the additive effect of these two cytotoxins is critical during Proteus infection. Since Pta is surface associated and important for the persistence of P. mirabilis in the host, it was selected as a vaccine candidate. Mice intranasally vaccinated with a site-directed (indicated by an asterisk) (S366A) mutant purified intact toxin (Pta*) or the passenger domain Pta-alpha*, each independently conjugated with cholera toxin (CT), had significantly lower bacterial counts in their kidneys ( P = 0.001) and spleens (P = 0.002) than mice that received CT alone. The serum immunoglobulin G levels correlated with protection (P = 0.03). This is the first report describing the in vivo cytotoxicity and antigenicity of an autotransporter in P. mirabilis and its use in vaccine development.


Subject(s)
Agglutinins/immunology , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Hemolysin Proteins/immunology , Proteus Infections/prevention & control , Proteus mirabilis , Urinary Tract Infections/microbiology , Urinary Tract Infections/prevention & control , Animals , Female , Kidney/microbiology , Kidney/pathology , Mice , Mice, Inbred CBA , Spleen/microbiology , Urinary Bladder/microbiology , Vaccination
14.
Antimicrob Agents Chemother ; 53(2): 393-400, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19015341

ABSTRACT

An engineered chitosan acetate bandage preparation (HemCon) is used as a hemostatic dressing, and its chemical structure suggests that it should also be antimicrobial. We previously showed that when a chitosan acetate bandage was applied to full-thickness excisional wounds in mice that had been infected with pathogenic bioluminescent bacteria (Pseudomonas aeruginosa, Proteus mirabilis, and Staphylococcus aureus), it was able to rapidly kill the bacteria and save the mice from developing fatal infections. Wound healing was also stimulated. In the present study, we asked whether a chitosan acetate bandage could act as a topical antimicrobial dressing when it was applied to third-degree burns in mice contaminated with two of these bacterial species (P. aeruginosa and P. mirabilis). Preliminary experiments established the length of burn time and the number of bacteria needed to produce fatal infections in untreated mice and established that the chitosan acetate bandage could adhere to the infected burn for up to 21 days. In the case of P. aeruginosa infections, the survival rate of mice treated with the chitosan acetate bandage was 73.3% (whereas the survival rate of mice treated with a nanocrystalline silver dressing was 27.3% [P = 0.0055] and that of untreated mice was 13.3% [P < 0.0002]). For P. mirabilis infections, the comparable survival rates were 66.7%, 62.5%, and 23.1% respectively. Quantitative bioluminescent signals showed that the chitosan acetate bandage effectively controlled the growth of bacteria in the burn and prevented the development of systemic sepsis, as shown by blood culture. These data suggest that chitosan acetate bandage is efficacious in preventing fatal burn infections.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/prevention & control , Bandages , Burns/complications , Chitosan , Administration, Topical , Animals , Bacterial Infections/microbiology , Burns/microbiology , Female , Luminescence , Mice , Mice, Inbred BALB C , Nanoparticles , Proteus Infections/microbiology , Proteus Infections/prevention & control , Proteus mirabilis , Silver/administration & dosage , Silver/therapeutic use , Survival Analysis
15.
J Urol ; 181(5): 2332-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19303096

ABSTRACT

PURPOSE: Proteus mirabilis is a common cause of urinary tract infection. We determined the role of Tamm-Horsfall protein as a host defense factor against the cystitis and pyelonephritis caused by P. mirabilis. MATERIALS AND METHODS: We generated Tamm-Horsfall protein gene knockout mice using homologous recombination. We introduced P. mirabilis transurethrally into the bladder of Tamm-Horsfall protein deficient (THP(-/-)) and genetically similar WT (THP(+/+)) mice. We cultured urine to quantitate the degree of bacteriuria. We examined bladders and kidneys grossly and histomorphometrically to determine the intensity of inflammation. RESULTS: THP(-/-) mice had more severe bacteriuria and cystitis than THP(+/+) mice. THP(-/-) mice had more pyelonephritic abscesses than THP(+/+) mice. The severity of histological pyelonephritis on semiquantitative histomorphometric analysis appeared to be greater in THP(-/-) mice. The difference between the 2 groups approached but did not attain statistical significance (p = 0.053). CONCLUSION: Tamm-Horsfall protein acts as a host defense factor against P. mirabilis induced urinary tract infection.


Subject(s)
Cystitis/pathology , Cystitis/prevention & control , Mucoproteins/genetics , Proteus Infections/prevention & control , Animals , Bacteriuria/genetics , Bacteriuria/physiopathology , Biopsy, Needle , Cystitis/genetics , Disease Models, Animal , Genetic Markers , Genetic Predisposition to Disease , Immunohistochemistry , Mice , Mice, Knockout , Mucoproteins/metabolism , Probability , Proteus Infections/genetics , Proteus Infections/physiopathology , Proteus mirabilis/pathogenicity , Random Allocation , Sensitivity and Specificity , Statistics, Nonparametric , Urinary Tract Infections/genetics , Urinary Tract Infections/pathology , Urinary Tract Infections/prevention & control , Uromodulin
16.
Pathol Biol (Paris) ; 57(3): e55-9, 2009 May.
Article in English | MEDLINE | ID: mdl-18456422

ABSTRACT

Emergence and dissemination of multiresistant strain of Proteus mirabilis have made infections treatment more difficult that this bacterium is responsible. The aim of this study is to determine the implication of the enzymatic mechanism and to describe the properties of ESBLs (extended spectrum beta-lactamases). A clinical strain of Proteus mirabilis SM514 isolated in the intensive care unit at the Military hospital in Tunisia during the period 2004 was found to be highly resistant to cephalosporins and penicilins. Cells sonicate of the isolate hydrolysed cefotaxime more efficiently than ceftriaxone and ceftazidime and had three beta-lactamases bands of approximate of isoelectric points (pI) of 5.4; 5.6 and superior to 7.6. The specific activities (AS) vary from 5.26 to 7.77U/mg of protein respectively for cefotaxime and the benzylpenicillin. These activities are inhibited by the clavulanic acid and the sulbactam. The values of the IC(50) are respectively 3.7 and 11.7muM. Only the beta-lactamases of pI 5.4 and superior to 7.6 hydrolyze the cefotaxime. Transformant produces the ESBLs of pI 5.4; 7.45 and greater than 7.6. The genes coding for this enzymes are carried by a transferable plasmids.


Subject(s)
Plasmids , Proteus mirabilis/enzymology , Proteus mirabilis/genetics , beta-Lactamases/genetics , Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Cross Infection/prevention & control , Drug Resistance, Bacterial , Drug Resistance, Multiple , Hospitals, Military , Humans , Intensive Care Units , Microbial Sensitivity Tests , Penicillins/pharmacology , Proteus Infections/epidemiology , Proteus Infections/prevention & control , Proteus Infections/transmission , Proteus mirabilis/drug effects , Tunisia , beta-Lactamase Inhibitors , beta-Lactamases/drug effects , beta-Lactamases/metabolism
17.
mSphere ; 4(1)2019 02 27.
Article in English | MEDLINE | ID: mdl-30814316

ABSTRACT

Proteus mirabilis is a common cause of catheter-associated urinary tract infection (CAUTI) and secondary bacteremia, which are frequently polymicrobial. We previously utilized transposon insertion-site sequencing (Tn-Seq) to identify novel fitness factors for colonization of the catheterized urinary tract during single-species and polymicrobial infection, revealing numerous metabolic pathways that may contribute to P. mirabilis fitness regardless of the presence of other cocolonizing organisms. One such "core" fitness factor was d-serine utilization. In this study, we generated isogenic mutants in d-serine dehydratase (dsdA), d-serine permease (dsdX), and the divergently transcribed activator of the operon (dsdC) to characterize d-serine utilization in P. mirabilis and explore the contribution of this pathway to fitness during single-species and polymicrobial infection. P. mirabilis was capable of utilizing either d- or l-serine as a sole carbon or nitrogen source, and dsdA, dsdX, and dsdC were each specifically required for d-serine degradation. This capability was highly conserved among P. mirabilis isolates, although not universal among uropathogens: Escherichia coli and Morganella morganii utilized d-serine, while Providencia stuartii and Enterococcus faecalis did not. d-Serine utilization did not contribute to P. mirabilis growth in urine ex vivo during a 6-h time course but significantly contributed to fitness during single-species and polymicrobial CAUTI during a 96-h time course, regardless of d-serine utilization by the coinfecting isolate. d-Serine utilization also contributed to secondary bacteremia during CAUTI as well as survival in a direct bacteremia model. Thus, we propose d-serine utilization as a core fitness factor in P. mirabilis and a possible target for disruption of infection.IMPORTANCE Urinary tract infections are among the most common health care-associated infections worldwide, the majority of which involve a urinary catheter (CAUTI). Our recent investigation of CAUTIs in nursing home residents identified Proteus mirabilis, Enterococcus species, and Escherichia coli as the three most common organisms. These infections are also often polymicrobial, and we identified Morganella morganii, Enterococcus species, and Providencia stuartii as being more prevalent during polymicrobial CAUTI than single-species infection. Our research therefore focuses on identifying "core" fitness factors that are highly conserved in P. mirabilis and that contribute to infection regardless of the presence of these other organisms. In this study, we determined that the ability to degrade d-serine, the most abundant d-amino acid in urine and serum, strongly contributes to P. mirabilis fitness within the urinary tract, even when competing for nutrients with another organism. d-Serine uptake and degradation therefore represent potential targets for disruption of P. mirabilis infections.


Subject(s)
Catheter-Related Infections/microbiology , Coinfection , Genetic Fitness , Proteus mirabilis/enzymology , Serine/metabolism , Urinary Tract Infections/microbiology , Animals , Female , Hydro-Lyases/genetics , Mice , Mutation , Operon , Proteus Infections/prevention & control , Proteus mirabilis/genetics
18.
Methods Mol Biol ; 2021: 201-215, 2019.
Article in English | MEDLINE | ID: mdl-31309507

ABSTRACT

Proteus mirabilis is a major cause of complicated urinary tract infections (UTIs). P. mirabilis' urease activity hydrolyzes urea and raises urine pH levels, which can catalyze bladder and kidney stone formation. This urolithiasis leads to harder-to-treat infections, possible urinary blockage, and subsequent septicemia. Development of a mucosal vaccine against P. mirabilis urinary tract infections is critical to protect against this potentially deadly infection process. Here, we describe the methodology necessary to produce a vaccine candidate conjugated to cholera toxin, administer the vaccine via the intranasal route, and test efficacy in a murine transurethral P. mirabilis infection model.


Subject(s)
Antibodies, Bacterial/metabolism , Immunotoxins/administration & dosage , Proteus Infections/prevention & control , Proteus mirabilis/immunology , Urinary Tract Infections/prevention & control , Administration, Intranasal , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Disease Models, Animal , Female , Humans , Immunotoxins/immunology , Mice , Mice, Inbred CBA , Vaccination
19.
J Hosp Infect ; 103(1): 55-63, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30802524

ABSTRACT

BACKGROUND: Catheter-associated urinary tract infections (CAUTIs) are among the most common hospital-acquired infections, leading to increased morbidity and mortality. A major reason for this is that urinary catheters are not yet capable of preventing CAUTIs. AIM: To develop an anti-infective urinary catheter. METHODS: An efficient silver-polytetrafluoroethylene (Ag-PTFE) nanocomposite coating was deposited on whole silicone catheters, and two in-vitro bladder models were designed to test antibacterial (against Escherichia coli) and anti-encrustation (against Proteus mirabilis) performances. Each model was challenged with two different concentrations of bacterial suspension. FINDINGS: Compared with uncoated catheters, coated catheters significantly inhibited bacterial migration and biofilm formation on the external catheter surfaces. The time to develop bacteriuria was an average of 1.8 days vs 4 days and 6 days vs 41 days when the urethral meatus was infected with 106 and 102 cells/mL, respectively. For anti-encrustation tests, the coated catheter significantly resisted encrustation, although it did not strongly inhibit the increases in bacterial density and urinary pH. The time to blockage, which was found to be independent of the initial bacterial concentration in the bladder, was extended from 36.2±1.1 h (uncoated) to 89.5±3.54 h (coated) following bacterial contamination with 103 cells/mL in the bladder. Moreover, the coated catheter exhibited excellent biocompatibility with L929 fibroblast cells. CONCLUSION: Ag-PTFE coated Foley catheters should undergo further clinical trials to determine their ability to prevent CAUTIs during catheterization.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Nanocomposites , Polytetrafluoroethylene/pharmacology , Proteus mirabilis/drug effects , Silver/pharmacology , Urinary Catheters , Bacteriuria/prevention & control , Catheter-Related Infections/prevention & control , Escherichia coli Infections/prevention & control , Humans , Models, Theoretical , Proteus Infections/prevention & control , Time Factors , Urinary Tract Infections/prevention & control
20.
Mol Immunol ; 96: 88-97, 2018 04.
Article in English | MEDLINE | ID: mdl-29525454

ABSTRACT

Proteus mirabilis is a common pathogen in urinary tract infections (UTIs). There is no vaccine against P. mirabilis, thus a novel multi-peptide vaccine of MrpA, UcaA and Pta factors of P. mirabilis we designed and a mice model was used to evaluate its efficacy in combination with AddaVax adjuvant. According to the bioinformatics studies, 7 fragments of MrpA (31-75, 112-146), UcaA (68-117, 132-156) and Pta (210-265, 340-400, 496-570) with B and T cell epitope regions were selected for fusion construction. Mice subcutaneously vaccinated with the fusion MrpA.Pta.UcaA induced a significant increase in serum and mucosal IgG and IgA responses. The fusion also showed a significant induction in cellular responses (Th1 and Th2). The addition of AddaVax to fusion and the mixture of MrpA, UcaA, and Pta (MUP) improved the humoral and cellular responses, especially the IgG2a and IFN-γ (Th1 responses) levels. Fusion with and without AddaVax and MUP + AddaVax could maintain significant humoral responses until 6 months after the first vaccine dose. All vaccine combinations with and without adjuvant showed high effectiveness in the protection of the bladder and kidney against experimental UTI; this could be attributed to the significant humoral and cellular responses. The present study suggests that the AddaVax-based vaccine formulations especially the fusion Pta.MrpA.UcaA admixed with AddaVax as potential vaccine candidates for protection against P. mirabilis. Furthermore, AddaVax could be considered as an effective adjuvant in designing other vaccines against UTI pathogens.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bacterial Vaccines/immunology , Proteus Infections/prevention & control , Squalene/immunology , Urinary Tract Infections/prevention & control , Animals , Bacterial Vaccines/pharmacology , Disease Models, Animal , Mice , Polysorbates/pharmacology , Proteus mirabilis , Squalene/pharmacology , Urinary Tract Infections/microbiology , Vaccines, Subunit
SELECTION OF CITATIONS
SEARCH DETAIL