Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(4): e2213363120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36652479

ABSTRACT

With the emergence of antibiotic-resistant bacteria, innovative approaches are needed for the treatment of urinary tract infections. Boosting antimicrobial peptide expression may provide an alternative to antibiotics. Here, we developed reporter cell lines and performed a high-throughput screen of clinically used drugs to identify compounds that boost ribonuclease 4 and 7 expression (RNase 4 and 7), peptides that have antimicrobial activity against antibiotic-resistant uropathogens. This screen identified histone deacetylase (HDAC) inhibitors as effective RNase 4 and RNase 7 inducers. Validation studies in primary human kidney and bladder cells confirmed pan-HDAC inhibitors as well as the HDAC class I inhibitor, MS-275, induce RNase 4 and RNase 7 to protect human kidney and bladder cells from uropathogenic Escherichia coli. When we administered MS-275 to mice, RNase 4 and 7 expression increased and mice were protected from acute transurethral E. coli challenge. In support of this mechanism, MS-275 treatment increased acetylated histone H3 binding to the RNASE4 and RNASE7 promoters. Overexpression and knockdown of HDAC class I proteins identified HDAC3 as a primary regulator of RNase 4 and 7. These results demonstrate the protective effects of enhancing RNase 4 and RNase 7, opening the door to repurposing medications as antibiotic conserving therapeutics for urinary tract infection.


Subject(s)
Histone Deacetylase Inhibitors , Urinary Tract Infections , Humans , Mice , Animals , Histone Deacetylase Inhibitors/pharmacology , Escherichia coli/metabolism , Drug Repositioning , Ribonucleases/metabolism , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology , Anti-Bacterial Agents
2.
Biomacromolecules ; 22(7): 2910-2920, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34085824

ABSTRACT

Antibiotic-resistant Gram-negative bacteria are emergent pathogens, causing millions of infections worldwide. While there are several classes of antibiotics that are effective against Gram-positive bacteria, the outer membrane (OM) of Gram-negative bacteria excludes high-molecular-weight hydrophobic antibiotics, making these species intrinsically resistant to several classes of antibiotics, including polyketides, aminocoumarins, and macrolides. The overuse of antibiotics such as ß-lactams has also promoted the spread of resistance genes throughout Gram-negative bacteria, including the production of extended spectrum ß-lactamases (ESBLs). The combination of innate and acquired resistance makes it extremely challenging to identify antibiotics that are effective against Gram-negative bacteria. In this study, we have demonstrated the synergistic effect of outer membrane-permeable cationic polyurethanes with rifampicin, a polyketide that would otherwise be excluded by the OM, on different strains of E. coli, including a clinically isolated uropathogenic multidrug-resistant (MDR) E. coli. Rifampicin combined with a low-dose treatment of a cationic polyurethane reduced the MIC in E. coli of rifampicin by up to 64-fold. The compositions of cationic polyurethanes were designed to have low hemolysis and low cell cytotoxicity while maintaining high antibacterial activity. Our results demonstrate the potential to rescue the large number of available OM-excluded antibiotics to target normally resistant Gram-negative bacteria via synergistic action with these cationic polyurethanes, acting as a novel antibiotic adjuvant class.


Subject(s)
Escherichia coli , Rifampin , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria , Microbial Sensitivity Tests , Polyurethanes , Rifampin/pharmacology
3.
Am J Physiol Renal Physiol ; 320(5): F972-F983, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33818125

ABSTRACT

Antimicrobial peptides are essential host defense mechanisms that prevent urinary tract infections. Recent studies have demonstrated that peptides in the ribonuclease A superfamily have antimicrobial activity against uropathogens and protect the urinary tract from uropathogenic Escherichia coli (UPEC). Little is known about the antibacterial function or expression of ribonuclease 4 (RNase 4) in the human urinary tract. Here, we show that full-length recombinant RNase 4 peptide and synthetic amino-terminal RNase 4 peptide fragment have antibacterial activity against UPEC and multidrug-resistant (MDR)-UPEC. RNASE4 transcript expression was detected in human kidney and bladder tissue using quantitative real-time PCR. Immunostaining or in situ hybridization localized RNase 4 expression to proximal tubules, principal and intercalated cells in the kidney's collecting duct, and the bladder urothelium. Urinary RNase 4 concentrations were quantified in healthy controls and females with a history of urinary tract infection. Compared with controls, urinary RNase 4 concentrations were significantly lower in females with a history of urinary tract infection. When RNase 4 was neutralized in human urine or silenced in vitro using siRNA, urinary UPEC replication or attachment to and invasion of urothelial and kidney medullary cells increased. These data show that RNase 4 has antibacterial activity against UPEC, is expressed in the human urinary tract, and can contribute to host defense against urinary tract infections.NEW & NOTEWORTHY Ribonuclease 4 (RNase 4) is a newly identified host defense peptide in the human kidney and bladder. RNase 4 kills uropathogenic Escherichia coli (UPEC) and multidrug-resistant UPEC. RNase 4 prevents invasive UPEC infection and suppressed RNase 4 expression may be a risk factor for more severe or recurrent urinary tract infection.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Kidney/enzymology , Ribonucleases/metabolism , Urinary Bladder/enzymology , Adolescent , Antimicrobial Cationic Peptides , Child , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Female , Gene Silencing , History, Ancient , History, Medieval , Humans , Kidney/metabolism , Real-Time Polymerase Chain Reaction , Ribonucleases/genetics , Ribonucleases/urine , Urinary Bladder/metabolism , Uropathogenic Escherichia coli , Urothelium/cytology
4.
J Am Soc Nephrol ; 30(8): 1385-1397, 2019 08.
Article in English | MEDLINE | ID: mdl-31239387

ABSTRACT

BACKGROUND: Evidence suggests that antimicrobial peptides, components of the innate immune response, protect the kidneys and bladder from bacterial challenge. We previously identified ribonuclease 7 (RNase 7) as a human antimicrobial peptide that has bactericidal activity against uropathogenic Escherichia coli (UPEC). Functional studies assessing RNase 7's contributions to urinary tract defense are limited. METHODS: To investigate RNase 7's role in preventing urinary tract infection (UTI), we quantified urinary RNase 7 concentrations in 29 girls and adolescents with a UTI history and 29 healthy female human controls. To assess RNase 7's antimicrobial activity in vitro in human urothelial cells, we used siRNA to silence urothelial RNase 7 production and retroviral constructs to stably overexpress RNase 7; we then evaluated UPEC's ability to bind and invade these cells. For RNase 7 in vivo studies, we developed humanized RNase 7 transgenic mice, subjected them to experimental UTI, and enumerated UPEC burden in the urine, bladder, and kidneys. RESULTS: Compared with controls, study participants with a UTI history had 1.5-fold lower urinary RNase 7 concentrations. When RNase 7 was silenced in vitro, the percentage of UPEC binding or invading human urothelial cells increased; when cells overexpressed RNase 7, UPEC attachment and invasion decreased. In the transgenic mice, we detected RNase 7 expression in the kidney's intercalated cells and bladder urothelium. RNase 7 humanized mice exhibited marked protection from UPEC. CONCLUSIONS: These findings provide evidence that RNase 7 has a role in kidney and bladder host defense against UPEC and establish a foundation for investigating RNase 7 as a UTI prognostic marker or nonantibiotic-based therapy.


Subject(s)
Escherichia coli Infections/enzymology , Kidney/enzymology , Ribonucleases/genetics , Urinary Bladder/enzymology , Urinary Tract Infections/enzymology , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli , Adolescent , Animals , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/genetics , Child , Child, Preschool , Female , Gene Silencing , Humans , Immunity, Innate , Infant , Kidney/microbiology , Male , Mice , Mice, Transgenic , Phenotype , Prognosis , Urinary Bladder/microbiology , Urothelium/metabolism , Urothelium/pathology , Young Adult
5.
J Clin Invest ; 128(12): 5634-5646, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30418175

ABSTRACT

People with diabetes mellitus have increased infection risk. With diabetes, urinary tract infection (UTI) is more common and has worse outcomes. Here, we investigate how diabetes and insulin resistance impact the kidney's innate defenses and urine sterility. We report that type 2 diabetic mice have increased UTI risk. Moreover, insulin-resistant prediabetic mice have increased UTI susceptibility, independent of hyperglycemia or glucosuria. To identify how insulin resistance affects renal antimicrobial defenses, we genetically deleted the insulin receptor in the kidney's collecting tubules and intercalated cells. Intercalated cells, located within collecting tubules, contribute to epithelial defenses by acidifying the urine and secreting antimicrobial peptides (AMPs) into the urinary stream. Collecting duct and intercalated cell-specific insulin receptor deletion did not impact urine acidification, suppressed downstream insulin-mediated targets and AMP expression, and increased UTI susceptibility. Specifically, insulin receptor-mediated signaling regulates AMPs, including lipocalin 2 and ribonuclease 4, via phosphatidylinositol-3-kinase signaling. These data suggest that insulin signaling plays a critical role in renal antibacterial defenses.


Subject(s)
Bacterial Infections/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Kidney Tubules, Collecting/metabolism , Receptor, Insulin/metabolism , Signal Transduction , Urinary Tract Infections/metabolism , Animals , Bacterial Infections/genetics , Bacterial Infections/microbiology , Bacterial Infections/pathology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/microbiology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/pathology , Kidney Tubules, Collecting/microbiology , Kidney Tubules, Collecting/pathology , Mice , Mice, Mutant Strains , Receptor, Insulin/genetics , Urinary Tract Infections/genetics , Urinary Tract Infections/pathology , alpha-Defensins/genetics , alpha-Defensins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...