Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
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
2.
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
3.
PLoS One ; 17(7): e0270067, 2022.
Article in English | MEDLINE | ID: mdl-35857780

ABSTRACT

MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression at the post-transcriptional level. miRNAs have been found in urine and have shown diagnostic potential in human nephropathies. Here, we aimed to characterize, for the first time, the feline urinary miRNAome and explore the use of urinary miRNA profiles as non-invasive biomarkers for feline pyelonephritis (PN). Thirty-eight cats were included in a prospective case-control study and classified in five groups: healthy Control cats (n = 11), cats with PN (n = 10), cats with subclinical bacteriuria or cystitis (SB/C, n = 5), cats with ureteral obstruction (n = 7) and cats with chronic kidney disease (n = 5). By small RNA sequencing we identified 212 miRNAs in cat urine, including annotated (n = 137) and putative novel (n = 75) miRNAs. The 15 most highly abundant urinary miRNAs accounted for nearly 71% of all detected miRNAs, most of which were previously identified in feline kidney. Ninety-nine differentially abundant (DA) miRNAs were identified when comparing Control cats to cats with urological conditions and 102 DA miRNAs when comparing PN to other urological conditions. Tissue clustering analysis revealed that the majority of urine samples clustered close to kidney, which confirm the likely cellular origin of the secreted urinary miRNAs. Relevant DA miRNAs were verified by quantitative real-time PCR (qPCR). Eighteen miRNAs discriminated Control cats from cats with a urological condition. Of those, seven miRNAs were DA by both RNAseq and qPCR methods between Control and PN cats (miR-125b-5p, miR-27a-3p, miR-21-5p, miR-27b-3p, miR-125a-5p, miR-17-5p and miR-23a-3p) or DA between Control and SB/C cats (miR-125b-5p). Six additional miRNAs (miR-30b-5p, miR-30c, miR-30e-5p, miR-27a-3p, miR-27b-39 and miR-222) relevant for discriminating PN from other urological conditions were identified by qPCR alone (n = 4) or by both methods (n = 2) (P<0.05). This panel of 13 miRNAs has potential as non-invasive urinary biomarkers for diagnostic of PN and other urological conditions in cats.


Subject(s)
MicroRNAs , Pyelonephritis , Renal Insufficiency, Chronic , Animals , Biomarkers/urine , Case-Control Studies , Cats , Humans , MicroRNAs/metabolism , Pyelonephritis/diagnosis , Pyelonephritis/genetics , Pyelonephritis/veterinary
4.
Nat Rev Urol ; 19(7): 419-437, 2022 07.
Article in English | MEDLINE | ID: mdl-35732832

ABSTRACT

Innovative solutions are needed for the treatment of bacterial infections, and a range of antibacterial molecules have been explored as alternatives to antibiotics. A different approach is to investigate the immune system of the host for new ways of making the antibacterial defence more efficient. However, the immune system has a dual role as protector and cause of disease: in addition to being protective, increasing evidence shows that innate immune responses can become excessive and cause acute symptoms and tissue pathology during infection. This role of innate immunity in disease suggests that the immune system should be targeted therapeutically, to inhibit over-reactivity. The ultimate goal is to develop therapies that selectively attenuate destructive immune response cascades, while augmenting the protective antimicrobial defence but such treatment options have remained underexplored, owing to the molecular proximity of the protective and destructive effects of the immune response. The concept of innate immunomodulation therapy has been developed successfully in urinary tract infections, based on detailed studies of innate immune activation and disease pathogenesis. Effective, disease-specific, immunomodulatory strategies have been developed by targeting specific immune response regulators including key transcription factors. In acute pyelonephritis, targeting interferon regulatory factor 7 using small interfering RNA or treatment with antimicrobial peptide cathelicidin was protective and, in acute cystitis, targeting overactive effector molecules such as IL-1ß, MMP7, COX2, cAMP and the pain-sensing receptor NK1R has been successful in vivo. Furthermore, other UTI treatment strategies, such as inhibiting bacterial adhesion and vaccination, have also shown promise.


Subject(s)
Cystitis , Pyelonephritis , Urinary Tract Infections , Anti-Bacterial Agents/therapeutic use , Cystitis/drug therapy , Humans , Immunomodulation , Pyelonephritis/drug therapy , Pyelonephritis/genetics , Pyelonephritis/microbiology , Urinary Tract Infections/drug therapy
5.
Am J Physiol Cell Physiol ; 321(1): C134-C146, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33979212

ABSTRACT

The local environment forces a selection of bacteria that might invade the urinary tract, allowing only the most virulent to access the kidney. Quite similar to the diet in setting the stage for the gut microbiome, renal function determines the conditions for bacteria-host interaction in the urinary tract. In the kidney, the term local environment or microenvironment is completely justified because the environment literally changes within a few micrometers. The precise composition of the urine is a function of the epithelium lining the microdomain, and the microenvironment in the kidney shows more variation in the content of nutrients, ion composition, osmolality, and pH than any other site of bacteria-host interaction. This review will cover some of the aspects of bacterial-host interaction in this unique setting and how uropathogenic bacteria can alter the condition for bacteria-host interaction. There will be a particular focus on the recent findings regarding how bacteria specifically trigger host paracrine signaling, via release of extracellular ATP and activation of P2 purinergic receptors. These finding will be discussed from the perspective of severe urinary tract infections, including pyelonephritis and urosepsis.


Subject(s)
Escherichia coli Infections/genetics , Escherichia coli Proteins/genetics , Hemolysin Proteins/genetics , Pyelonephritis/genetics , Receptors, Purinergic P2/genetics , Sepsis/genetics , Urinary Tract Infections/genetics , Uropathogenic Escherichia coli/genetics , Adenosine Triphosphate/metabolism , Anoctamin-1/genetics , Anoctamin-1/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli Proteins/metabolism , Gene Expression Regulation , Hemolysin Proteins/metabolism , Host-Pathogen Interactions/genetics , Humans , Hydrogen-Ion Concentration , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Paracrine Communication , Pyelonephritis/metabolism , Pyelonephritis/microbiology , Pyelonephritis/pathology , Receptors, Purinergic P2/metabolism , Sepsis/metabolism , Sepsis/microbiology , Sepsis/pathology , Signal Transduction , Urinary Tract Infections/metabolism , Urinary Tract Infections/microbiology , Urinary Tract Infections/pathology , Uropathogenic Escherichia coli/growth & development , Uropathogenic Escherichia coli/metabolism , Uropathogenic Escherichia coli/pathogenicity
6.
Article in English | MEDLINE | ID: mdl-33010454

ABSTRACT

BACKGROUND: Several studies revealed alterations of single sphingolipid species, such as chain length-specific ceramides, in plasma and serum of patients with kidney diseases. Here, we investigated whether such alterations occur in kidney tissue from patients and mice suffering from renal fibrosis, the common endpoint of chronic kidney diseases. METHODS: Human fibrotic kidney samples were collected from nephrectomy specimens with hydronephrosis and/or pyelonephritis. Healthy parts from tumor nephrectomies served as nonfibrotic controls. Mouse fibrotic kidney samples were collected from male C57BL/6J mice treated with an adenine-rich diet for 14 days or were subjected to 7 days of unilateral ureteral obstruction (UUO). Kidneys of untreated mice and contralateral kidneys (UUO) served as respective controls. Sphingolipid levels were detected by LC-MS/MS. Fibrotic markers were analyzed by TaqMan® analysis and immunohistology. RESULTS: Very long-chain ceramides Cer d18:1/24:0 and Cer d18:1/24:1 were significantly downregulated in both fibrotic human kidney cortex and fibrotic murine kidney compared to respective control samples. These effects correlate with upregulation of COL1α1, COL3α1 and αSMA expression in fibrotic human kidney cortex and fibrotic mouse kidney. CONCLUSION: We have shown that very long-chain ceramides Cer d18:1/24:0 and Cer d18:1/24:1 are consistently downregulated in fibrotic kidney samples from human and mouse. Our findings support the use of in vivo murine models as appropriate translational means to understand the involvement of ceramides in human kidney diseases. In addition, our study raises interesting questions about the possible manipulation of ceramide metabolism to prevent progression of fibrosis and the use of ceramides as potential biomarkers of chronic kidney disease.


Subject(s)
Ceramides/metabolism , Hydronephrosis/metabolism , Pyelonephritis/metabolism , Sphingolipids/metabolism , Ureteral Obstruction/metabolism , Actins/genetics , Actins/metabolism , Adenine/administration & dosage , Aged , Animals , Biomarkers/metabolism , Ceramides/classification , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Collagen Type III/genetics , Collagen Type III/metabolism , Disease Models, Animal , Female , Fibrosis , Gene Expression Regulation , Humans , Hydronephrosis/chemically induced , Hydronephrosis/genetics , Hydronephrosis/pathology , Kidney/metabolism , Kidney/pathology , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Middle Aged , Pyelonephritis/chemically induced , Pyelonephritis/genetics , Pyelonephritis/pathology , Sphingolipids/classification , Ureteral Obstruction/genetics , Ureteral Obstruction/pathology
7.
Am J Physiol Renal Physiol ; 318(6): F1441-F1453, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32390512

ABSTRACT

Carbonic anhydrase II knockout (Car2-/-) mice have depleted numbers of renal intercalated cells, which are increasingly recognized to be innate immune effectors. We compared pyelonephritis susceptibility following reciprocal renal transplantations between Car2-/- and wild-type mice. We examined the effect of pharmacological CA suppression using acetazolamide in an experimental murine model of urinary tract infection. Car2-/- versus wild-type mice were compared for differences in renal innate immunity. In our transplant scheme, mice lacking CA-II in the kidney had increased pyelonephritis risk. Mice treated with acetazolamide had lower kidney bacterial burdens at 6 h postinfection, which appeared to be due to tubular flow from diuresis because comparable results were obtained when furosemide was substituted for acetazolamide. Isolated Car2-/- kidney cells enriched for intercalated cells demonstrated altered intercalated cell innate immune gene expression, notably increased calgizzarin and insulin receptor expression. Intercalated cell number and function along with renal tubular flow are determinants of pyelonephritis risk.


Subject(s)
Acetazolamide/pharmacology , Carbonic Anhydrase II/deficiency , Carbonic Anhydrase Inhibitors/pharmacology , Escherichia coli Infections/prevention & control , Kidney/drug effects , Pyelonephritis/prevention & control , Urinary Tract Infections/prevention & control , Acidosis/enzymology , Acidosis/genetics , Animals , Carbonic Anhydrase II/antagonists & inhibitors , Carbonic Anhydrase II/genetics , Disease Models, Animal , Escherichia coli Infections/enzymology , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Immunity, Innate , Kidney/enzymology , Kidney/immunology , Kidney/microbiology , Kidney Transplantation , Mice, Inbred C57BL , Mice, Knockout , Pyelonephritis/enzymology , Pyelonephritis/genetics , Pyelonephritis/microbiology , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , S100 Proteins/genetics , S100 Proteins/metabolism , Urinary Tract Infections/enzymology , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/pathogenicity
8.
Nat Rev Nephrol ; 16(2): 112-128, 2020 02.
Article in English | MEDLINE | ID: mdl-31831877

ABSTRACT

Advances in single-cell technologies are transforming our understanding of cellular identity. For instance, the application of single-cell RNA sequencing and mass cytometry technologies to the study of immune cell populations in blood, secondary lymphoid organs and the renal tract is helping researchers to map the complex immune landscape within the kidney, define cell ontogeny and understand the relationship of kidney-resident immune cells with their circulating counterparts. These studies also provide insights into the interactions of immune cell populations with neighbouring epithelial and endothelial cells in health, and across a range of kidney diseases and cancer. These data have translational potential and will aid the identification of drug targets and enable better prediction of off-target effects. The application of single-cell technologies to clinical renal biopsy samples, or even cells within urine, will improve diagnostic accuracy and assist with personalized prognostication for patients with various kidney diseases. A comparison of immune cell types in peripheral blood and secondary lymphoid organs in healthy individuals and in patients with systemic autoimmune diseases that affect the kidney will also help to unravel the mechanisms that underpin the breakdown in self-tolerance and propagation of autoimmune responses. Together, these immune cell atlases have the potential to transform nephrology.


Subject(s)
Acute Kidney Injury/immunology , Autoimmune Diseases/immunology , Immune System/cytology , Lymphocytes/metabolism , Renal Insufficiency, Chronic/immunology , Single-Cell Analysis , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Flow Cytometry , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Kidney Transplantation , Leukocytes/immunology , Leukocytes/metabolism , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Lymphocytes/immunology , Pyelonephritis/genetics , Pyelonephritis/immunology , Pyelonephritis/metabolism , RNA-Seq , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Sequence Analysis, RNA
9.
Molecules ; 25(1)2019 Dec 25.
Article in English | MEDLINE | ID: mdl-31881666

ABSTRACT

Nowadays, saliva is a subject of growing scientific interest because of its definite advantages as diagnostic medium. The aim of our study was to investigate the diagnostic potential and reliability of messenger RNAs (mRNAs) of selected genes-interleukin-6 (IL-6), matrix metalloproteinase-8 (MMP-8) and glutathione synthetase (GSS)-as salivary markers in children with diagnosed pyelonephritis and to correlate their levels with typical urine para-clinical indicators of the disease. Analysis of the mRNA levels for IL-6, MMP-8 and GSS in 28 children hospitalized with the diagnosis of pyelonephritis was conducted applying the method of quantitative reverse transcription polymerase chain reaction (RT-qPCR). In the study group (n = 28), IL-6 mRNA levels demonstrated 64-fold increase (p < 0.001). MMP-8 and GSS mRNA levels were increased in 12 samples in patients with pyelonephritis 3.27 (p < 0.01) and 1.94 (p < 0.001) times, respectively. We found a strong and significant correlation (p < 0.001) between the investigated mRNA for IL-6 and MMP-8, IL-6 and GSS, MMP-8 and GSS. Moderate degree of correlation was established between IL-6 and the typical para-clinical indicator of leucocytes (0.43, p < 0.05) and between GSS and leucocytes (0.54, p < 0.01). Salivary IL-6, MMP-8 and GSS mRNA levels in combination with urine test analysis could be useful diagnostic tool for the very distributed disorder of pyelonephritis in childhood.


Subject(s)
Glutathione Synthase/genetics , Interleukin-6/genetics , Matrix Metalloproteinase 8/genetics , Pyelonephritis/genetics , Saliva/metabolism , Biomarkers/urine , Child , Child, Preschool , Female , Glutathione Synthase/metabolism , Humans , Interleukin-6/metabolism , Male , Matrix Metalloproteinase 8/metabolism , Pyelonephritis/diagnosis , Pyelonephritis/urine , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Georgian Med News ; (288): 44-48, 2019 Mar.
Article in Russian | MEDLINE | ID: mdl-31101774

ABSTRACT

Goal of the study - estimation of the transforming growth factor ß1 and monocyte chemoattractant protein-1 (MCP-1) in infants with pyelonephritis. It was found that pyelonephritis against the background of vesicoureteral reflux is accompanied by the high activity of the inflammatory process and increase of serum monocyte chemoattractant protein-1 levels (2.5 times higher than in children with primary pyelonephritis). A significant relationship between the content of monocytic chemoattractant protein-1 and C-reactive protein (rxy=0,66, р<0,01) in young children with pyelonephritis against the background of reflux was established. Pyelonephritis against the background of vesicoureteral reflux in young children is accompanied by an increase in the content of serum transforming growth factor ß1 (TGF-ß1) which is 2,8 times higher, in comparison with patients with primary pyelonephritis. The profibrotic marker increased as the disease activity increased. In children of early age with pielonefritis on the background of vesicoureteral reflux and the presence of genotype C-509С and T+869Т (65,02±6,74%) transforming growth factor ß1 (TGF ß1) gene the probability of developing the disease is 4.48 times higher, compared to genotype of T-509Т+869С and 3.03 times higher compared to genotype S-509Т and T+869С.


Subject(s)
Chemokine CCL2 , Pyelonephritis , Transforming Growth Factor beta1 , Vesico-Ureteral Reflux , Biomarkers , C-Reactive Protein , Chemokine CCL2/genetics , Child , Child, Preschool , Genotype , Humans , Infant , Pyelonephritis/diagnosis , Pyelonephritis/genetics , Transforming Growth Factor beta1/genetics
12.
Am J Pathol ; 189(6): 1201-1211, 2019 06.
Article in English | MEDLINE | ID: mdl-30926332

ABSTRACT

Severe urinary tract infections are commonly caused by sub-strains of Escherichia coli secreting the pore-forming virulence factor α-hemolysin (HlyA). Repeated or severe cases of pyelonephritis can cause renal scarring that subsequently can lead to progressive failure. We have previously demonstrated that HlyA releases cellular ATP directly through its membrane pore and that acute HlyA-induced cell damage is completely prevented by blocking ATP signaling. Local ATP signaling and P2X7 receptor activation play a key role in the development of tissue fibrosis. This study investigated the effect of P2X7 receptors on infection-induced renal scarring in a murine model of pyelonephritis. Pyelonephritis was induced by injecting 100 million HlyA-producing, uropathogenic E. coli into the urinary bladder of BALB/cJ mice. A similar degree of pyelonephritis and mortality was confirmed at day 5 after infection in P2X7+/+ and P2X7-/- mice. Fibrosis was first observed 2 weeks after infection, and the data clearly demonstrated that P2X7-/- mice and mice exposed to the P2X7 antagonist, brillian blue G, show markedly less renal fibrosis 14 days after infection compared with controls (P < 0.001). Immunohistochemistry revealed comparable early neutrophil infiltration in the renal cortex from P2X7+/+ and P2X7-/- mice. Interestingly, lack of P2X7 receptors resulted in diminished macrophage infiltration and reduced neutrophil clearance in the cortex of P2X7-/- mice. Hence, this study suggests the P2X7 receptor to be an appealing antifibrotic target after renal infections.


Subject(s)
Escherichia coli Proteins/metabolism , Hemolysin Proteins/metabolism , Kidney/metabolism , Pyelonephritis , Receptors, Purinergic P2X7/deficiency , Uropathogenic Escherichia coli , Animals , Fibrosis , Kidney/microbiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Pyelonephritis/genetics , Pyelonephritis/metabolism , Pyelonephritis/microbiology , Pyelonephritis/prevention & control , Receptors, Purinergic P2X7/metabolism , Uropathogenic Escherichia coli/metabolism , Uropathogenic Escherichia coli/pathogenicity
13.
Bull Exp Biol Med ; 166(3): 334-338, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30627903

ABSTRACT

Activities of MMP-2 and MMP-9 in the cytoplasm and mitochondria of kidney cells were evaluated on the models of acute renal pathologies: pyelonephritis, rhabdomyolysis, and ischemia/reperfusion of the kidney. In acute pyelonephritis, a significant increase in the level of MMP-2 and MMP-9 in kidney cells and the appearance of mitochondrial MMP-2 isoform with a lower molecular weight, but still exhibiting proteolytic activity were observed. A direct correlation between the level of MMP-2 and MMP-9 in the kidney and the severity of inflammation in pyelonephritis was revealed. Obviously, the appearance of active protease in the mitochondria of the kidney cells could have an impact on their functioning and, generally, on the fate of renal cells in this pathology.


Subject(s)
Bacterial Infections/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Mitochondria/genetics , Pyelonephritis/genetics , Reperfusion Injury/genetics , Rhabdomyolysis/genetics , Acute Disease , Animals , Animals, Outbred Strains , Bacterial Infections/enzymology , Bacterial Infections/pathology , Disease Models, Animal , Epithelial Cells , Gene Expression Regulation , Isoenzymes/genetics , Isoenzymes/metabolism , Kidney/enzymology , Kidney/pathology , Kidney/surgery , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mitochondria/enzymology , Mitochondria/pathology , Nephrectomy/methods , Pyelonephritis/enzymology , Pyelonephritis/pathology , Rats , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Rhabdomyolysis/enzymology , Rhabdomyolysis/pathology , Severity of Illness Index
14.
Nephrology (Carlton) ; 24(4): 464-471, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29577511

ABSTRACT

AIM: The aim of this study was to determine whether a correlation exists between interleukin-8 receptor polymorphisms and urinary tract infection (UTI) susceptibility. METHODS: We systematically searched electronic databases including PubMed, Embase, China National Knowledge Infrastructure, and Web of Science up to 5 November 2017 to select appropriate studies that focused on C-X-C chemokine receptor type 1 and/or 2 (CXCR1, CXCR2) polymorphisms with susceptibility to UTI. Eight case-control studies including 2085 patients with UTI and 2012 controls were enrolled in this study. Seven studies of CXCR1 rs2234671 and two studies of rs3138086 were included in the meta-analyses. Pooled odds ratio (OR) and corresponding 95% confidence interval (CI) were synthesized using fixed-effects or random-effects model according to heterogeneity. RESULTS: No significant correlations were found between CXCR1 rs2234671 and rs3138086 polymorphisms and UTI susceptibility. However, subgroup analysis showed that rs2234671 was associated with an increased risk of UTI under allelic comparisons (C vs. G, OR = 1.95, 95% CI = 1.07-3.55), heterozygous model (GC vs. GG, OR = 1.93, 95% CI = 1.06-3.50), and dominant model (GC + CC vs. GG, OR = 1.98, 95% CI = 1.07-3.69) in children, especially in paediatric patients with acute pyelonephritis (allelic, OR = 2.43, 95% CI = 1.28-4.60; heterozygous, OR = 2.40, 95% CI = 1.24-4.62; dominant, OR = 2.48, 95% CI = 1.26-4.88). Furthermore, these results remained the same after eliminating paediatric patients with vesicoureteral reflux. CONCLUSION: CXCR1 rs2234671 polymorphism might be associated with an increased risk of UTI in children.


Subject(s)
Polymorphism, Single Nucleotide , Pyelonephritis/genetics , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8B/genetics , Urinary Tract Infections/genetics , Age Factors , Case-Control Studies , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Heterozygote , Homozygote , Humans , Phenotype , Protective Factors , Pyelonephritis/diagnosis , Pyelonephritis/prevention & control , Risk Factors , Urinary Tract Infections/diagnosis , Urinary Tract Infections/prevention & control
16.
Sci Rep ; 8(1): 6535, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29695842

ABSTRACT

Metal limitation is a common situation during infection and can have profound effects on the pathogen's success. In this report, we examine the role of zinc limitation in the expression of a virulence factor in uropathogenic Escherichia coli. The pyelonephritis isolate J96 carries two hlyCABD operons that encode the RTX toxin α-hemolysin. While the coding regions of both operons are largely conserved, the upstream sequences, including the promoters, are unrelated. We show here that the two hlyCABD operons are differently regulated. The hly II operon is efficiently silenced in the presence of zinc and highly expressed when zinc is limited. In contrast, the hly I operon does not respond to zinc limitation. Genetic studies reveal that zinc-responsive regulation of the hly II operon is controlled by the Zur metalloregulatory protein. A Zur binding site was identified in the promoter sequence of the hly II operon, and we observe direct binding of Zur to this promoter region. Moreover, we find that Zur regulation of the hly II operon modulates the ability of E. coli J96 to induce a cytotoxic response in host cell lines in culture. Our report constitutes the first description of the involvement of the zinc-sensing protein Zur in directly modulating the expression of a virulence factor in bacteria.


Subject(s)
Hemolysin Proteins/genetics , Uropathogenic Escherichia coli/genetics , Virulence Factors/genetics , Virulence/genetics , Zinc/metabolism , Bacterial Proteins/genetics , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/genetics , Operon/genetics , Promoter Regions, Genetic/genetics , Pyelonephritis/genetics
17.
Acta Paediatr ; 107(2): 339-346, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29028278

ABSTRACT

AIM: We studied the functional polymorphisms of intercellular adhesion molecule-1 (ICAM-1) and toll-like receptor-4 (TLR-4) genes and risk of acute pyelonephritis (APN) in children attending Assiut University Children's Hospitals, Egypt, from 2011 to 2015. METHODS: Urinary tract infections (UTIs) were diagnosed in 380 children: 98 had APN and 282 had lower UTIs. Four single-nucleotide polymorphisms in ICAM-1 and TLR-4 genes were genotyped in all subjects: ICAM-1 rs1799969 Gly241Arg, ICAM-1 rs5498 Glu469Lys, TLR-4 rs4896791 Thr399Ile and TLR-4 rs4896790 Asp299Gly. RESULTS: Patients with APN were significantly more likely to have AA genotype of the ICAM-1 rs5498 (1462 A/G) polymorphism (p = 0.04) than children with lower UTIs and the TLR-4 Asp299Gly GG genotype (p = 0.002) and G allele (p = 0.006) than healthy controls. The association with the ICAM-1 Glu469Lys (1462A/G) was less evident. The GG genotype was associated with a modest relative risk of 1.4 (p = 0.1) of developing APN, but was not an independent odds ratio, at 1.2 (p = 0.48). CONCLUSION: Functional variants in ICAM-1 and TLR-4 genes were increasingly common in children with febrile UTIs with renal parenchymal involvement, but the ICAM-1 Glu469Lys (1462A/G) association was less evident. TLR4 Asp299Gly might independently increase renal parenchymal infection rather than renal scarring.


Subject(s)
Intercellular Adhesion Molecule-1/genetics , Pyelonephritis/genetics , Toll-Like Receptor 4/genetics , Urinary Tract Infections/genetics , Adolescent , Case-Control Studies , Child , Child, Preschool , Escherichia coli/isolation & purification , Female , Fever/etiology , Genotype , Humans , Kidney/pathology , Male , Parenchymal Tissue/pathology , Polymorphism, Genetic , Pyelonephritis/etiology , Urinary Tract Infections/complications , Urine/microbiology
18.
J Pediatr Urol ; 13(6): 593.e1-593.e10, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28716390

ABSTRACT

BACKGROUND: Acute pyelonephritis is associated with renal scarring in up to 30% of patients. Renal scarring may cause significant long-term morbidity. The pathogenesis of acute pyelonephritis remains unclear, although it involves interaction among uroepithelium, the immune system cells, and the locally produced cytokines. That some UTI-prone children develop acute pyelonephritis, and eventually renal parenchymal scarring, suggests a genetic role. Interleukin-6, interleukin-8, chemokine receptor-1 (CXCR1), and tumor necrosis factor-alpha (TNFα), the key regulators of the host immune responses, are proteins whose secretion is controlled by genes. We postulated that functional polymorphic variants of their genes might have a role in APN susceptibility. OBJECTIVES: We sought to investigate a possible association of the common functional polymorphisms in genes encoding IL-6, IL-8, CXCR1, and TNFα with the risk of APN in children. METHODS: Urine culture was used to diagnose 300 children with UTI, of mean age of 51.31 ± 37.4 months (2-180 months). 99Tc-DMSA scans diagnosed 86 children with APN. Follow-up scans identified new renal scars in 18 children. Six functional single-nucleotide polymorphisms (SNPs) in genes encoding IL-6, IL-8, CXCR1, and TNFα were genotyped in all subjects (IL-6 rs1800795 (-174G/C), IL-6 rs1800796 (-572G/C), IL-8 rs2227306 (781C/T), IL8 rs4073 (-251A/T), CXCR1 rs2234671 (2607G/C), and TNFα rs1800629 (-308G/A)). RESULTS: TT genotype of IL-8 -251A/T polymorphism was significantly higher in APN patients (26.7%) than those with lower UTI (11.7%, p = 0.01) and control individuals (12.2%, p = 0.002). T allele was significantly more common in APN than in lower UTI (p = 0.025) and was significantly more common in APN (46%) than in the controls (p = 0.001). Similarly, TT genotype of IL-8 781C/T polymorphism was significantly more common in APN patients (31.4%) than those with lower UTI (17.3%, p = 0.003) and the controls (14.3%, p = 0.001). T allele was significantly more common in APN (55%) than lower UTI (40%, p = 0.005) and controls (37%, p = 0.001). However, IL-8 -251A/T and +781C/T SNPs did not qualify as an independent risk for parenchymal infection (OR 1.9, 95% CI 0.68-2.6, p = 0.13 and OR 2.3, 95% CI 0.89-3.7, p = 0.091, respectively). Lower UTI did not differ from the controls. The frequency of the genotypes and alleles of IL-6, CXCR1, and TNFα SNPs did not differ significantly among the different groups of the study. CONCLUSION: IL-8 -251A/T and +781C/T SNPs are associated with susceptibility to renal parenchymal infection in children and could be implicated in APN risk. However, none of these variants could clearly and independently predict this risk.


Subject(s)
Cytokines/genetics , Polymorphism, Single Nucleotide , Pyelonephritis/genetics , Pyelonephritis/microbiology , Urinary Tract Infections/genetics , Acute Disease , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Female , Genetic Predisposition to Disease , Humans , Male , Risk Factors , Young Adult
19.
Clin Transplant ; 30(9): 1115-33, 2016 09.
Article in English | MEDLINE | ID: mdl-27352120

ABSTRACT

Differentiating acute pyelonephritis (APN) from acute rejection (AR) in renal allograft biopsies can sometimes be difficult because of overlapping clinical and histologic features, lack of positive urine cultures,and variable response to antibiotics. We wanted to study differential gene expression between AR and APN using biopsy tissue. Thirty-three biopsies were analyzed using NanoString multiplex platform and PCR (6 transplant baseline biopsies, 8 AR, 15 APN [8 culture positive, 7 culture negative], and 4 native pyelonephritis [NP]). Additional 22 biopsies were tested by PCR to validate the results. CXCL9, CXCL10, CXCL11, and IDO1 were the top differentially expressed genes, upregulated in AR. Lactoferrin (LTF) and CXCL1 were higher in APN and NP. No statistically significant difference in transcript levels was seen between culture-positive and culture-negative APN biopsies. Comparing the overall mRNA signature using Ingenuity pathway analysis, interferon-gamma emerged as the dominant upstream regulator in AR and allograft APN, but not in NP (which clustered separately). Our study suggests that chemokine pathways in graft APN may differ from NP and in fact resemble AR, due to a component of alloreactivity, resulting in variable response to antibiotic treatment. Therefore, cautious addition of steroids might help in resistant cases of graft APN.


Subject(s)
Biopsy/methods , Chemokines, CXC/genetics , Gene Expression Regulation , Graft Rejection/genetics , Kidney Transplantation/adverse effects , Kidney/pathology , Pyelonephritis/genetics , Adult , Aged , Allografts , Chemokines, CXC/biosynthesis , Female , Follow-Up Studies , Graft Rejection/metabolism , Graft Rejection/pathology , Humans , Male , Middle Aged , Polymerase Chain Reaction , Pyelonephritis/metabolism , Pyelonephritis/pathology , RNA/genetics , Retrospective Studies , Young Adult
20.
Sci Transl Med ; 8(336): 336ra59, 2016 04 27.
Article in English | MEDLINE | ID: mdl-27122612

ABSTRACT

Boosting innate immunity represents an important therapeutic alternative to antibiotics. However, the molecular selectivity of this approach is a major concern because innate immune responses often cause collateral tissue damage. We identify the transcription factor interferon regulatory factor 7 (IRF-7), a heterodimer partner of IRF-3, as a target for non-antibiotics-based therapy of bacterial infections. We found that the efficient and self-limiting innate immune response to bacterial infection relies on a tight balance between IRF-3 and IRF-7. Deletion of Irf3 resulted in overexpression of Irf7 and led to an IRF-7-driven hyperinflammatory phenotype, which was entirely prevented if Irf7 was deleted. We then identified a network of strongly up-regulated, IRF-7-dependent genes in Irf3(-/-) mice with kidney pathology, which was absent in Irf7(-/-) mice. IRF-3 and IRF-7 from infected kidney cell nuclear extracts were shown to bind OAS1, CCL5, and IFNB1 promoter oligonucleotides. These data are consistent in children with low IRF7 expression in the blood: attenuating IRF7 promoter polymorphisms (rs3758650-T and rs10902179-G) negatively associated with recurrent pyelonephritis. Finally, we identified IRF-7 as a target for immunomodulatory therapy. Administering liposomal Irf7 siRNA to Irf3(-/-) mice suppressed mucosal IRF-7 expression, and the mice were protected against infection and renal tissue damage. These findings offer a response to the classical but unresolved question of "good versus bad inflammation" and identify IRF7 as a therapeutic target for protection against bacterial infection.


Subject(s)
Bacterial Infections/immunology , Immunity, Innate/physiology , Interferon Regulatory Factor-7/metabolism , Animals , Bacterial Infections/metabolism , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans , Immunity, Innate/genetics , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/genetics , Kidney/metabolism , Mice , Mice, Inbred C57BL , Pyelonephritis/genetics , Pyelonephritis/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...