Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 249
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732074

ABSTRACT

Early diagnosis of infections in young infants remains a clinical challenge. Young infants are particularly vulnerable to infection, and it is often difficult to clinically distinguish between bacterial and viral infections. Urinary tract infection (UTI) is the most common bacterial infection in young infants, and the incidence of associated bacteremia has decreased in the recent decades. Host RNA expression signatures have shown great promise for distinguishing bacterial from viral infections in young infants. This prospective study included 121 young infants admitted to four pediatric emergency care departments in the capital region of Denmark due to symptoms of infection. We collected whole blood samples and performed differential gene expression analysis. Further, we tested the classification performance of a two-gene host RNA expression signature approaching clinical implementation. Several genes were differentially expressed between young infants with UTI without bacteremia and viral infection. However, limited immunological response was detected in UTI without bacteremia compared to a more pronounced response in viral infection. The performance of the two-gene signature was limited, especially in cases of UTI without bloodstream involvement. Our results indicate a need for further investigation and consideration of UTI in young infants before implementing host RNA expression signatures in clinical practice.


Subject(s)
Urinary Tract Infections , Humans , Urinary Tract Infections/genetics , Infant , Prospective Studies , Female , Male , Transcriptome , Infant, Newborn , Gene Expression Profiling/methods , Bacteremia/genetics , RNA/genetics , Virus Diseases/genetics
2.
Life Sci Alliance ; 7(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38580392

ABSTRACT

Antimicrobial peptides (AMPs) are host defense effectors with potent neutralizing and immunomodulatory functions against invasive pathogens. The AMPs α-Defensin 1-3/DEFA1A3 participate in innate immune responses and influence patient outcomes in various diseases. DNA copy-number variations in DEFA1A3 have been associated with severity and outcomes in infectious diseases including urinary tract infections (UTIs). Specifically, children with lower DNA copy numbers were more susceptible to UTIs. The mechanism of action by which α-Defensin 1-3/DEFA1A3 copy-number variations lead to UTI susceptibility remains to be explored. In this study, we use a previously characterized transgenic knock-in of the human DEFA1A3 gene mouse to dissect α-Defensin 1-3 gene dose-dependent antimicrobial and immunomodulatory roles during uropathogenic Escherichia coli (UPEC) UTI. We elucidate the relationship between kidney neutrophil- and collecting duct intercalated cell-derived α-Defensin 1-3/DEFA1A3 expression and UTI. We further describe cooperative effects between α-Defensin 1-3 and other AMPs that potentiate the neutralizing activity against UPEC. Cumulatively, we demonstrate that DEFA1A3 directly protects against UPEC meanwhile impacting pro-inflammatory innate immune responses in a gene dosage-dependent manner.


Subject(s)
Urinary Tract Infections , alpha-Defensins , Animals , Humans , Mice , alpha-Defensins/genetics , DNA , Gene Dosage , Immunity, Innate/genetics , Kidney/metabolism , Peptides, Cyclic/genetics , Urinary Tract Infections/genetics , Urinary Tract Infections/metabolism
3.
Hum Antibodies ; 32(1): 9-18, 2024.
Article in English | MEDLINE | ID: mdl-38339925

ABSTRACT

BACKGROUND: Urinary tract infections (UTIs) are currently posing a worldwide health concern by affecting millions of people. The genetic variant rs2234671 in the CXCR1-interleukin-8 receptor is closely related to a raised UTI risk. OBJECTIVES: In this work, the impact of CXCR1 (rs2234671) on UTI individuals was examined. METHODS: The demographic features of 30 recurrent UTI patients and 20 controls were thoroughly investigated. Bacterial isolation and identification were performed by the implementation of cultural and biochemical methods. DNA extraction, purification of all samples from both patients and healthy people, and IL-8 rs2234671 (C/G) SNP genotyping using T-ARMS-PCR were performed. The significance of the results was evaluated by carrying out a statistical analysis. FINDINGS: The patient's average age was 34.63 ± 11.44 years, and controls averaged 30.30 ± 8.59 years (P= 0.156). No significant gender difference existed (P= 0.804). Escherichia coli (63.3%) was predominant, followed by Proteus mirabilis (26.7%), Enterococcus faecalis (23.3%), Klebsiella pneumoniae (10.0%), and Pseudomonas aeruginosa (20.0%). No significant association was found between bacterial species frequency, age, or sex. From the CXCR1 (rs2234671) frequency comparison, a higher GG genotype incidence in UTI patients than controls was extracted (26.7% vs. 15.0%), though not statistically significant. Risk analysis revealed that GG homozygous and C/G heterozygous genotypes were not UTI risk factors (OR = 2.47 and OR = 1.85, respectively). Moreover, the allele frequencies displayed no significant difference between the patients and controls (G allele: 66.7% vs. 66.7%; C allele: 33.3% vs. 33.3%). MAIN CONCLUSIONS: Although no significant association between CXCR1 (rs2234671) and UTI was found, the GG genotype may point to the increasing probability of UTI risk. Additional research is required to confirm and expand these conclusions.


Subject(s)
Urinary Tract Infections , Adult , Humans , Middle Aged , Young Adult , Alleles , Gene Frequency/genetics , Genotype , Risk Factors , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology
4.
Epigenomics ; 15(19): 961-964, 2023 10.
Article in English | MEDLINE | ID: mdl-37905413

ABSTRACT

Tweetable abstract Exploring uropathogenic E. coli-induced epigenetic changes in uroepithelial cells contributing to recurrent UTIs and potential therapeutic strategies. Understanding these mechanisms could inform novel UTI interventions.


Subject(s)
Escherichia coli Infections , Urinary Tract Infections , Uropathogenic Escherichia coli , Humans , Escherichia coli Infections/drug therapy , Urinary Tract Infections/genetics , Urinary Tract Infections/drug therapy , Epigenesis, Genetic , Epigenomics , Uropathogenic Escherichia coli/genetics
5.
Neurol Sci ; 44(12): 4491-4498, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37452996

ABSTRACT

Infantile hypotonia with psychomotor retardation and characteristic facies 1 (IHPRF1) is caused by biallelic mutations in the NALCN gene, the major ion channel responsible for the background Na + conduction in neurons. Through whole-exome sequencing (WES), we report three novel homozygous variants in three families, including c.1434 + 1G > A, c.3269G > A, and c.2648G > T, which are confirmed and segregated by Sanger sequencing. Consequently, intron 12's highly conserved splice donor location is disrupted by the pathogenic c.1434 + 1G > A variation, most likely causing the protein to degrade through nonsense-mediated decay (NMD). Subsequently, a premature stop codon is thus generated at amino acid 1090 of the protein as a result of the pathogenic c.3269G > A; p.W1090* variation, resulting in NMD or truncated protein production. Lastly, the missense mutation c.2648G > T; p.G883V can play a critical role in the interplay of functional domains. This study introduces recurrent urinary tract infections for the first time, broadening the phenotypic range of IHPRF1 syndrome in addition to the genotypic spectrum. This trait may result from insufficient bladder emptying, which may be related to the NALCN channelosome's function in background Na + conduction. This work advances knowledge about the molecular genetic underpinnings of IHPRF1 and introduces a novel phenotype through the widespread use of whole exome sequencing.


Subject(s)
Sodium Channels , Urinary Tract Infections , Humans , Sodium Channels/genetics , Sodium Channels/metabolism , Ion Channels/genetics , Membrane Proteins/genetics , Phenotype , Mutation, Missense , Syndrome , Urinary Tract Infections/genetics , Mutation/genetics
6.
BMC Med Genomics ; 16(1): 122, 2023 06 05.
Article in English | MEDLINE | ID: mdl-37277784

ABSTRACT

BACKGROUND: Some bladder-related diseases, such as bladder urinary tract infection (UTI) and bladder cancer (BCa), have significant six differences in incidence and prognosis. However, the molecular mechanisms underlying these sex differences are still not fully understood. Understanding the sex-biased differences in gene expression in normal bladder cells can help resolve these problems. METHODS: We first collected published single-cell RNA sequencing (scRNA-seq) data of normal human bladders from females and males to map the bladder transcriptomic landscape. Then, Gene Ontology (GO) analysis and gene set enrichment analysis (GSEA) were used to determine the significant pathways that changed in the specific cell populations. The Monocle2 package was performed to reconstruct the differentiation trajectories of fibroblasts. In addition, the scMetabolism package was used to analyze the metabolic activity at the single-cell level, and the SCENIC package was used to analyze the regulatory network. RESULTS: In total, 27,437 cells passed stringent quality control, and eight main cell types in human bladder were identified according to classical markers. Sex-based differential gene expression profiles were mainly observed in human bladder urothelial cells, fibroblasts, B cells, and T cells. We found that urothelial cells in males demonstrated a higher growth rate. Moreover, female fibroblasts produced more extracellular matrix, including seven collagen genes that may mediate BCa progression. Furthermore, the results showed that B cells in female bladders exhibited more B-cell activated signals and a higher expression of immunoglobulin genes. We also found that T cells in female bladders exhibited more T-cell activated signals. These different biological functions and properties of these cell populations may correlate with sex differences in UTI and BCa, and result in different disease processes and outcomes. CONCLUSIONS: Our study provides reasonable insights for further studies of sex-based physiological and pathological disparities in the human bladder, which will contribute to the understanding of epidemiological differences in UTI and BCa.


Subject(s)
Urinary Bladder Neoplasms , Urinary Bladder , Urinary Tract Infections , Humans , Prospective Studies , Urinary Bladder/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Tract Infections/genetics , Single-Cell Analysis , Gene Expression Regulation , Sequence Analysis, RNA
7.
Mol Biol Rep ; 50(7): 5817-5826, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37219671

ABSTRACT

BACKGROUND: Proteus mirabilis is a Gram-negative bacteria most noted for its involvement with catheter-associated urinary tract infections. It is also known for its multicellular migration over solid surfaces, referred to as 'swarming motility'. Here we analyzed the genomic sequences of two P. mirabilis isolates, designated K38 and K39, which exhibit varied swarming ability. METHODS AND RESULTS: The isolates genomes were sequenced using Illumina NextSeq sequencer, resulting in about 3.94 Mbp, with a GC content of 38.6%, genomes. Genomes were subjected for in silico comparative investigation. We revealed that, despite a difference in swarming motility, the isolates showed high genomic relatedness (up to 100% ANI similarity), suggesting that one of the isolates probably originated from the other. CONCLUSIONS: The genomic sequences will allow us to investigate the mechanism driving this intriguing phenotypic heterogeneity between closely related P. mirabilis isolates. Phenotypic heterogeneity is an adaptive strategy of bacterial cells to several environmental pressures. It is also an important factor related to their pathogenesis. Therefore, the availability of these genomic sequences will facilitate studies that focus on the host-pathogen interactions during catheter-associated urinary tract infections.


Subject(s)
Proteus Infections , Urinary Tract Infections , Humans , Proteus mirabilis/genetics , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology , Clone Cells , Proteus Infections/microbiology
8.
Infect Immun ; 91(2): e0055922, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36651775

ABSTRACT

Healthcare-acquired infections are a leading cause of disease in patients that are hospitalized or in long-term-care facilities. Klebsiella pneumoniae (Kp) is a leading cause of bacteremia, pneumonia, and urinary tract infections in these settings. Previous studies have established that the ter operon, a genetic locus that confers tellurite oxide (K2TeO3) resistance, is associated with infection in colonized patients. Rather than enhancing fitness during infection, the ter operon increases Kp fitness during gut colonization; however, the biologically relevant function of this operon is unknown. First, using a murine model of urinary tract infection, we demonstrate a novel role for the ter operon protein TerC as a bladder fitness factor. To further characterize TerC, we explored a variety of functions, including resistance to metal-induced stress, resistance to radical oxygen species-induced stress, and growth on specific sugars, all of which were independent of TerC. Then, using well-defined experimental guidelines, we determined that TerC is necessary for tolerance to ofloxacin, polymyxin B, and cetylpyridinium chloride. We used an ordered transposon library constructed in a Kp strain lacking the ter operon to identify the genes that are required to resist K2TeO3-induced and polymyxin B-induced stress, which suggested that K2TeO3-induced stress is experienced at the bacterial cell envelope. Finally, we confirmed that K2TeO3 disrupts the Kp cell envelope, though these effects are independent of ter. Collectively, the results from these studies indicate a novel role for the ter operon as a stress tolerance factor, thereby explaining its role in enhancing fitness in the gut and bladder.


Subject(s)
Bacteremia , Klebsiella Infections , Urinary Tract Infections , Humans , Animals , Mice , Klebsiella pneumoniae/genetics , Polymyxin B/pharmacology , Operon , Urinary Tract Infections/genetics , Bacteremia/genetics , Klebsiella Infections/microbiology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism
9.
J Med Life ; 15(8): 955-962, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36188660

ABSTRACT

This study aimed to assess the role of paternal genetics in the development of diabetic mellitus (DM) and determine the impact of DM on the urinary system by investigating the percentage of patients with urinary tract infection (UTI). The study included 100 people with DM; their ages ranged from 5 to 83 years. The DM and blood sugar levels were diagnosed clinically and at a laboratory in Al-Zahra Teaching Hospital and the outpatient clinics. The age, gender, and causes of DM and the family history of diabetes were reported. Isolation and identification of bacterial species were made depending on culture media and biochemical tests. The average age of patients was 47.7±5.5, and most of them were female (67%). The incidence of DM increased with age, and the main cause of DM was likely to be a genetic predisposition (family history), where 32% of patients appeared to have a positive family history and the presence of DM in both parents or only the mother had a significant role in increasing the genetic predisposition of developing DM. Among the non-genetic causes of DM, the most common was exposure to sudden psychological or nervous shock (41%). Obesity also had an important role in the development of diabetes, and also pregnancy and smoking. Moreover, 66% of patients with type 2 DM and all with type 1 DM suffered from UTIs. The main causative agents were E. coli (60%) and Proteus spp. (13%). The majority of patients suffering from UTIs (73%) were females. In conclusion, type 2 DM is the most common, especially in females, and increases with age. The main cause of DM was family genetic predisposition and sudden shocks. The current study also showed that most diabetic patients suffered from UTIs, especially females, and the main causes of UTI inflammation are E. coli isolates.


Subject(s)
Diabetes Mellitus, Type 2 , Urinary Tract Infections , Adolescent , Adult , Aged , Aged, 80 and over , Blood Glucose , Child , Child, Preschool , Culture Media , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Escherichia coli , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Parents , Pregnancy , Urinary Tract Infections/complications , Urinary Tract Infections/epidemiology , Urinary Tract Infections/genetics , Young Adult
10.
J Biomed Sci ; 29(1): 66, 2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36068602

ABSTRACT

BACKGROUND: Extraintestinal Escherichia coli (E. coli) causing urinary tract infections (UTIs), and often referred to as uropathogenic E. coli (UPEC), are a major contributor to the morbidity of UTIs and associated healthcare costs. UPEC possess several virulence factors (VFs) for infecting and injuring the host. We studied the papG allele distribution, and its association with other VF genes and phylogenetic groups, amongst 836 UPEC and fecal isolates from reproductive age women. RESULTS: The papGII gene was highly prevalent amongst pyelonephritis isolates (68%), whilst the majority, albeit smaller proportion, of cystitis isolates (31%) harboured the papGIII gene. Among the pyelonephritis and cystitis isolates, papG positive isolates on average had higher VF gene scores, and were more likely to belong to phylogenetic group B2, than their negative counterparts. This was mostly due to the contribution of papGII isolates, which on average contained more VF genes than their papGIII counterparts, irrespective of the uro-clinical syndrome. However, the papGII isolates from the pyelonephritis cohort had higher VF gene scores than the cystitis ones, suggesting presence of possible papGII clones with differing inferred virulence potential. Furthermore, papGII isolates were more likely to possess an intact pap gene operon than their papGIII counterparts. Also of note was the high proportion of isolates with the papGI allele which was not associated with other pap operon genes; and this finding has not been described before. CONCLUSIONS: The association of the papGII gene with several VF genes compared to the papGIII gene, appears to explain the abundance of these genes in pyelonephritis and cystitis isolates, respectively.


Subject(s)
Cystitis , Escherichia coli Infections , Pyelonephritis , Urinary Tract Infections , Uropathogenic Escherichia coli , Adhesins, Escherichia coli/genetics , Alleles , Cystitis/genetics , Escherichia coli Infections/epidemiology , Escherichia coli Infections/genetics , Female , Fimbriae Proteins/genetics , Humans , Phylogeny , Pyelonephritis/genetics , Urinary Tract Infections/genetics , Uropathogenic Escherichia coli/genetics , Virulence Factors/genetics
11.
Proc Natl Acad Sci U S A ; 119(40): e2206515119, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36161923

ABSTRACT

Antimicrobial peptides (AMPs) are critical to the protection of the urinary tract of humans and other animals from pathogenic microbial invasion. AMPs rapidly destroy pathogens by disrupting microbial membranes and/or augmenting or inhibiting the host immune system through a variety of signaling pathways. We have previously demonstrated that alpha-defensins 1-3 (DEFA1A3) are AMPs expressed in the epithelial cells of the human kidney collecting duct in response to uropathogens. We also demonstrated that DNA copy number variations in the DEFA1A3 locus are associated with UTI and pyelonephritis risk. Because DEFA1A3 is not expressed in mice, we utilized human DEFA1A3 gene transgenic mice (DEFA4/4) to further elucidate the biological relevance of this locus in the murine urinary tract. We demonstrate that the kidney transcriptional and translational expression pattern is similar in humans and the human gene transgenic mouse upon uropathogenic Escherichia coli (UPEC) stimulus in vitro and in vivo. We also demonstrate transgenic human DEFA4/4 gene mice are protected from UTI and pyelonephritis under various UPEC challenges. This study serves as the foundation to start the exploration of manipulating the DEFA1A3 locus and alpha-defensins 1-3 expression as a potential therapeutic target for UTIs and other infectious diseases.


Subject(s)
Escherichia coli Infections , Pyelonephritis , Urinary Tract Infections , Uropathogenic Escherichia coli , alpha-Defensins , Animals , DNA Copy Number Variations , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Genetic Loci , Humans , Mice , Mice, Transgenic , Pyelonephritis/genetics , Pyelonephritis/immunology , Pyelonephritis/microbiology , Urinary Tract/microbiology , Urinary Tract Infections/genetics , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , alpha-Defensins/genetics
12.
Indian J Med Res ; 155(3&4): 423-431, 2022 03.
Article in English | MEDLINE | ID: mdl-36124515

ABSTRACT

Background & objectives: Infections caused by vancomycin-resistant Enterococci are difficult to treat given the limited therapeutic alternatives. Different gene clusters are known to confer vancomycin resistance. vanA and vanB genes are transferable and are clinically relevant. This cross-sectional study aimed to identify the vancomycin-resistant genotypes in the strains causing urinary tract infection and also to test the in vitro efficacy of linezolid and pristinamycin against the vancomycin-resistant isolates. Methods: Antimicrobial resistance profile of 118 enterococcal isolates was evaluated. Minimum inhibitory concentration of vancomycin, teicoplanin and high-level gentamicin (HLG) was determined by micro broth dilution. The vancomycin-resistant isolates were tested against linezolid and pristinamycin by micro-broth dilution and E strip method. The presence of vancomycin-resistant genes was detected by multiplex polymerase chain reaction and was sequenced and analyzed. Results: Most commonly isolated species were Enterococcus faecalis (76.9%) and Enterococcus faecium (16.9%). It was found that 43 per cent of the isolates were resistant to HLG and 16.9 per cent to vancomycin. Higher resistance was seen against fluoroquinolones, erythromycin, tetracycline and ß-lactam drugs. However, 5.08 per cent strains were resistant to tigecycline. All vancomycin-resistant strains were sensitive to pristinamycin and one was resistant to linezolid. vanA and vanB gene were found in 15 and five isolates, respectively. The gene sequences were submitted to NCBI gene bank and accession numbers were obtained. Interpretation & conclusions: The present study showed prevalence of vanA and vanB genes carrying Enterococcus in a tertiary care centre in north India. The emergence of resistance against drugs such as tigecycline and linezolid is a topic of concern as it will be a therapeutic challenge for physicians.


Subject(s)
Urinary Tract Infections , Vancomycin-Resistant Enterococci , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cross-Sectional Studies , Erythromycin , Fluoroquinolones , Genotype , Gentamicins , Humans , Linezolid/therapeutic use , Pristinamycin , Teicoplanin , Tigecycline , Urinary Tract Infections/drug therapy , Urinary Tract Infections/epidemiology , Urinary Tract Infections/genetics , Vancomycin/therapeutic use , beta-Lactams
13.
Genes (Basel) ; 13(8)2022 08 06.
Article in English | MEDLINE | ID: mdl-36011308

ABSTRACT

The rapid emergence of multidrug-resistant (MDR) bacteria indisputably constitutes a major global health problem. Pathogenic Escherichia coli are listed among the most critical group of bacteria that require fast development of new antibiotics and innovative treatment strategies. Among harmful extraintestinal Enterobacteriaceae strains, uropathogenic E. coli (UPEC) pose a significant health threat. UPEC are considered the major causative factor of urinary tract infection (UTI), the second-most commonly diagnosed infectious disease in humans worldwide. UTI treatment places a substantial financial burden on healthcare systems. Most importantly, the misuse of antibiotics during treatment has caused selection of strains with the ability to acquire MDR via miscellaneous mechanisms resulting in gaining resistance against many commonly prescribed antibiotics like ampicillin, gentamicin, cotrimoxazole and quinolones. Mobile genetic elements (MGEs) such as transposons, integrons and conjugative plasmids are the major drivers in spreading resistance genes in UPEC. The co-occurrence of various bacterial evasion strategies involving MGEs and the SOS stress response system requires further research and can potentially lead to the discovery of new, much-awaited therapeutic targets. Here, we analyzed and summarized recent discoveries regarding the role, mechanisms, and perspectives of MDR in the pathogenicity of UPEC.


Subject(s)
Escherichia coli Infections , Urinary Tract Infections , Uropathogenic Escherichia coli , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli Infections/drug therapy , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Humans , Urinary Tract Infections/drug therapy , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/genetics
14.
mBio ; 13(3): e0103522, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35546538

ABSTRACT

More than half of women will experience a urinary tract infection (UTI), with uropathogenic Escherichia coli (UPEC) causing ~80% of uncomplicated cases. Iron acquisition systems are essential for uropathogenesis, and UPEC strains encode highly diverse iron acquisition systems, underlining their importance. However, a recent UPEC clinical isolate, HM7, lacks this diversity and instead encodes the synthesis pathway for a sole siderophore, enterobactin. To determine if HM7 possesses unidentified iron acquisition systems, we performed RNA sequencing under iron-limiting conditions and demonstrated that the ferric citrate uptake system (fecABCDE and fecIR) was highly upregulated. Importantly, there are high levels of citrate within urine, some of which is bound to iron, and the fec system is enriched in UPEC isolates compared to fecal strains. Therefore, we hypothesized that HM7 and other similar strains use the fec system to acquire iron in the host. Deletion of both enterobactin biosynthesis and ferric citrate uptake (ΔfecA/ΔentB) abrogates use of ferric citrate as an iron source, and fecA provides an advantage in human urine in the absence of enterobactin. However, in a UTI mouse model, fecA is a fitness factor independent of enterobactin production, likely due to the action of host lipocalin-2 chelating ferrienterobactin. These findings indicate that ferric citrate uptake is used as an iron source when siderophore efficacy is limited, such as in the host during UTI. Defining these novel compensatory mechanisms and understanding the nutritional hierarchy of preferred iron sources within the urinary tract are important in the search for new approaches to combat UTI. IMPORTANCE UPEC, the primary causative agent of uncomplicated UTI, is responsible for five billion dollars in health care costs in the United States each year. Rates of antibiotic resistance are on the rise; therefore, it is vital to understand the mechanisms of UPEC pathogenesis to uncover potential targets for novel therapeutics. Iron acquisition systems used to obtain iron from sequestered host sources are essential for UPEC survival during UTI and have been used as vaccine targets to prevent infection. This study reveals the ferric citrate uptake system is another important iron acquisition system that is highly enriched in UPEC strains. Ferric citrate uptake has not previously been associated with UPEC isolates, underlining the importance of the continued study of these strains to fully understand their mechanisms of pathogenesis.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Urinary Tract Infections , Uropathogenic Escherichia coli , Animals , Citric Acid/metabolism , Enterobactin/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Ferric Compounds , Humans , Iron/metabolism , Mice , Receptors, Cell Surface/metabolism , Siderophores/metabolism , Urinary Tract Infections/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
15.
Sci Rep ; 12(1): 2005, 2022 02 07.
Article in English | MEDLINE | ID: mdl-35132157

ABSTRACT

The inflammasome-associated proteins caspase-1, caspase-4 and NLRP3 have been emphasised to be essential in the host cell response during urinary tract infection (UTI) by regulating IL-1ß release. Our aim was to investigate how the inflammasome-associated proteins regulate the cell response of bladder epithelial cells during infection with uropathogenic Escherichia coli (UPEC). Human bladder epithelial cells (5637) and CRISPR/Cas9 generated caspase-1, caspase-4 and NLRP3 knockdown cells were stimulated with the UPEC strain CFT073. Using Olink proteomics and real time RT-PCR, we showed that caspase-1, caspase-4 and NLRP3 are vital for the expression of many inflammatory genes and proteins from bladder epithelial cells. When investigating the effect of inflammasome-associated proteins on neutrophils, we found that conditioned medium from UPEC-infected caspase-4 knockdown cells significantly increased phagocytosis of CFT073 and significantly decreased ROS production from neutrophils. In contrast, conditioned medium from UPEC-infected NLRP3 knockdown cells significantly decreased the phagocytosis of CFT073 and significantly increased the ROS production from neutrophils. In conclusion, we showed that the inflammasome-associated proteins contribute to the host cell response during UPEC infection.


Subject(s)
Caspase 1/physiology , Caspases, Initiator/physiology , Epithelial Cells/immunology , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Urinary Tract Infections/genetics , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli/immunology , Caspases, Initiator/metabolism , Cell Line , Epithelial Cells/metabolism , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Inflammation , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophils/metabolism , Phagocytosis , Reactive Oxygen Species/metabolism , Urinary Bladder/cytology
16.
PLoS One ; 17(1): e0262984, 2022.
Article in English | MEDLINE | ID: mdl-35077517

ABSTRACT

Carbapenems are the last resort antimicrobials for the treatment of extended spectrum ß-lactamases (ESBLs) producing Enterobacteriaceae. Emergence of carbapenems resistant group B2 uropathogenic E. coli (UPEC) is a major concern because of their high virulence. Prevalence of these enzymes and multidrug resistance (MDR) among B2 UPEC isolates from Iraqi outpatients with acute urinary tract infection (UTI) was evaluated in this research. Urine cultures were performed and the isolates were identified biochemically. Escherichia coli isolates were tested for phylogroup reference by quadraplex PCR, then B2 isolates were detected for antimicrobial resistance by disc diffusion test and carbapenemase genes by PCR. Escherichia coli was the most prevalent among Gram-negative isolates (66.6%) and B2 was the most detected phylogroup among E. coli isolates (33.9%). Most of B2 isolates showed high resistance rates to tested antimicrobials, especially ß-lactams with MDR revealed in 100% of them. Whereas, low resistance rates were noted against carbapenems, aminoglycosides and nitrofurantoin. Carbapenemase genes were detected in 76.3% of B2 isolates. Of which, blaOXA-48 was the most frequent (57.8%), followed by blaPER (47.3%), blaKPC (15.7%), blaVEB and blaVIM (10.5%, for each). Whereas, blaGES and blaIMP genes were not found. Coproduction of these genes occurred among 17 isolates. The combination of blaOXA-48 and blaPER was the most frequent (41.1%). All carbapenemase producing isolates were MDR. These results revealed high prevalence of carbapenemase genes and MDR among B2 UPEC recovered in this study. In the study area. it is strongly advised to use aminoglycosides and nitrofurantoin for empirical treatment of UPEC.


Subject(s)
Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial , Escherichia coli Infections/genetics , Outpatients , Urinary Tract Infections/genetics , Uropathogenic Escherichia coli , Virulence Factors/genetics , beta-Lactamases/genetics , Escherichia coli Infections/enzymology , Escherichia coli Infections/epidemiology , Female , Humans , Iraq/epidemiology , Male , Prevalence , Urinary Tract Infections/enzymology , Urinary Tract Infections/epidemiology , Uropathogenic Escherichia coli/enzymology , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/isolation & purification
17.
PLoS One ; 17(1): e0262952, 2022.
Article in English | MEDLINE | ID: mdl-35089940

ABSTRACT

The uropathogens is the main cause of urinary tract infection (UTI). The aim of the study was to isolate bacteria from urine samples of UTI patients and find out the susceptibility of isolated bacteria. Bacteria were identified using both conventional and molecular methods. Sanger sequence procedure used for 16S ribosomal RNA and phylogenetic analysis was performed using Molecular Evolutionary Genetics Analysis (MEGA-7) software. In this study, Escherichia coli, Klebsiella pneumonia, Staphylococcus were reported as 58, 28 and 14.0% respectively. Phylogenetic tree revealed that 99% of sample No. Ai (05) is closely related to E. coli to (NR 114042.1 E. coli strain NBRC 102203). Aii (23) is 99% similar to K. pneumoniae to (NR 117686.1 K. pneumonia strain DSM 30104) and 90% Bi (48) is closely linked to S. aureus to (NR 113956.1 S. aureus strain NBRC 100910). The antibiotic susceptibility of E. coli recorded highest resistance towards ampicillin (90%) and least resistant to ofloxacin (14%). Some of the other antibiotics such amoxicillin, ciprofloxacin, gentamicin, ceftazidime, cefuroxime and nitrofurantoin resistance were observed 86, 62, 24, 55, 48 and 35% respectively. The cefuroxime showed the highest antibiotic resistance against K. pneumoniae with 85% followed by amoxicillin, ciprofloxacin, gentamicin, ceftazidime, ampicillin and nitrofurantoin resulted in 60, 45, 67, 70, 75 and 30% respectively. The resistance of S. aureus against erythromycin, cefuroxime and ampicillin were found with 72%. The resistance against amoxicillin, gentamicin, ceftazidime and ceftriaxone found 57, 43, 43 and 15% respectively. Phylogenetic analysis shows that sequences are closely related with the reference sequences and E. coli is the dominant bacteria among UTI patients and is resistant to the commercially available antibiotics.


Subject(s)
Bacteria , Bacterial Infections , Drug Resistance, Bacterial/genetics , Phylogeny , Urinary Tract Infections , Anti-Bacterial Agents/pharmacology , Bacteria/genetics , Bacteria/growth & development , Bacteria/isolation & purification , Bacterial Infections/genetics , Bacterial Infections/microbiology , Female , Humans , Male , Microbial Sensitivity Tests , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology
18.
mBio ; 13(1): e0351921, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35038925

ABSTRACT

Many antibiotic resistant uropathogenic Escherichia coli (UPEC) strains belong to clones defined by their multilocus sequence type (ST), with ST131 being the most dominant. Although we have a good understanding of resistance development to fluoroquinolones and third-generation cephalosporins by ST131, our understanding of the virulence repertoire that has contributed to its global dissemination is limited. Here we show that the genes encoding Afa/Dr fimbriae, a group of adhesins strongly associated with UPEC that cause gestational pyelonephritis and recurrent cystitis, are found in approximately one third of all ST131 strains. Sequence comparison of the AfaE adhesin protein revealed a unique allelic variant carried by 82.9% of afa-positive ST131 strains. We identify the afa regulatory region as a hotspot for the integration of insertion sequence (IS) elements, all but one of which alter afa transcription. Close investigation demonstrated that the integration of an IS1 element in the afa regulatory region leads to increased expression of Afa/Dr fimbriae, promoting enhanced adhesion to kidney epithelial cells and suggesting a mechanism for altered virulence. Finally, we provide evidence for a more widespread impact of IS1 on ST131 genome evolution, suggesting that IS dynamics contribute to strain level microevolution that impacts ST131 fitness. IMPORTANCE E. coli ST131 is the most common antibiotic resistant UPEC clone associated with human urinary tract and bloodstream infections. Understanding the features of ST131 that have driven its global dissemination remains a critical priority if we are to counter its increasing antibiotic resistance. Here, we utilized a large collection of ST131 isolates to investigate the prevalence, regulation, and function of Afa/Dr fimbriae, a well-characterized UPEC colonization and virulence factor. We show that the afa genes are found frequently in ST131 and demonstrate how the integration of IS elements in the afa regulatory region modulates Afa expression, presenting an example of altered virulence capacity. We also exploit a curated set of ST131 genomes to map the integration of the antibiotic resistance-associated IS1 element in the ST131 pangenome, providing evidence for its widespread impact on ST131 genome evolution.


Subject(s)
Drug Resistance, Multiple, Bacterial , Escherichia coli Infections , Urinary Tract Infections , Uropathogenic Escherichia coli , Humans , Adhesins, Bacterial/metabolism , Anti-Bacterial Agents/metabolism , Clone Cells , DNA Transposable Elements , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli Infections/genetics , Urinary Tract Infections/genetics , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/pathogenicity , Virulence/genetics
19.
Infect Immun ; 90(2): e0027521, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34871042

ABSTRACT

Uropathogenic Escherichia coli (UPEC) causes the majority of uncomplicated urinary tract infections (UTI), which affect nearly half of women worldwide. Many UPEC strains carry an annotated intimin-like adhesin (ila) locus in their genome related to a well-characterized virulence factor in diarrheagenic E. coli pathotypes. Its role in UPEC uropathogenesis, however, remains unknown. In prototype UPEC strain CFT073, there is an ila locus that contains three predicted intimin-like genes, sinH, sinI, and ratA. We used in silico approaches to determine the phylogeny and genomic distribution of this locus among uropathogens. We found that the currently annotated intimin locus-encoded proteins in CFT073 are more closely related to invasin proteins found in Salmonella. Deletion of the individual sinH, sinI, and ratA genes did not result in measurable effects on growth, biofilm formation, or motility in vitro. On average, sinH was more highly expressed in clinical strains during active human UTI than in human urine ex vivo. Unexpectedly, we found that strains lacking this ila locus had increased adherence to bladder cells in vitro, coupled with a decrease in bladder cell invasion and death. The sinH mutant displayed a significant fitness defect in the murine model of ascending UTI, including reduced inflammation in the bladder. These data confirmed an inhibitory role in bladder cell adherence to facilitate invasion and inflammation; therefore, the ila locus should be termed invasin-like rather than intimin-like. Collectively, our data suggest that loss of this locus mediates measurable interactions with bladder cells in vitro and contributes to fitness during UTI.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Urinary Tract Infections , Uropathogenic Escherichia coli , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Animals , Escherichia coli Infections/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Genomic Islands/genetics , Humans , Inflammation/genetics , Male , Mice , Urinary Tract Infections/genetics , Urothelium
20.
J Mol Med (Berl) ; 100(1): 101-113, 2022 01.
Article in English | MEDLINE | ID: mdl-34651203

ABSTRACT

Infections are common in patients with diabetes, but increasing antibiotic resistance hampers successful bacterial clearance and calls for alternative treatment strategies. Hypoxia-inducible factor 1 (HIF-1) is known to influence the innate immune defense and could therefore serve as a possible target. However, the impact of high glucose on HIF-1 has received little attention and merits closer investigation. Here, we show that higher levels of proinflammatory cytokines and CAMP, encoding for the antimicrobial peptide cathelicidin, LL-37, correlate with HIF-1 in type 2 diabetic patients. Chemical activation of HIF-1 further enhanced LL-37, IL-1ß, and IL-8 in human uroepithelial cells exposed to high glucose. Moreover, HIF-1 activation of transurethrally infected diabetic mice resulted in lower bacterial load. Drugs activating HIF-1 could therefore in the future potentially have a therapeutic role in clearing bacteria in diabetic patients with infections where antibiotic treatment failed. KEY MESSAGES: • Mohanty et al. "HIF-1 mediated activation of antimicrobial peptide LL-37 in type 2 diabetic patients." • Our study highlights induction of the antimicrobial peptide, LL-37, and strengthening of the innate immunity through hypoxia-inducible factor 1 (HIF-1) in diabetes. • Our key observations are: 1. HIF-1 activation increased LL-37 expression in human urothelial cells treated with high glucose. In line with that, we demonstrated that patients with type 2 diabetes living at high altitude had increased levels of the LL-37. 2. HIF-1 activation increased IL-1ß and IL-8 in human uroepithelial cells treated with high glucose concentration. 3. Pharmacological activation of HIF-1 decreased bacterial load in the urinary bladder of mice with hereditary diabetes. • We conclude that enhancing HIF-1 may along with antibiotics in the future contribute to the treatment in selected patient groups where traditional therapy is not possible.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 2/immunology , Escherichia coli Infections/immunology , Hypoxia-Inducible Factor 1/immunology , Urinary Tract Infections/immunology , Adult , Aged , Aged, 80 and over , Animals , Cytokines/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Escherichia coli Infections/genetics , Female , Humans , Hypoxia-Inducible Factor 1/genetics , Male , Mice , Middle Aged , Urinary Tract Infections/genetics , Urothelium/cytology , Cathelicidins
SELECTION OF CITATIONS
SEARCH DETAIL
...