Human neutrophil peptides 1-3 protect the murine urinary tract from uropathogenic Escherichia coli challenge
Edited by Michael Zasloff, Georgetown University Medical Center, Washington, DC; received April 19, 2022; accepted August 8, 2022 by Editorial Board Member Stephen J. Benkovic
Significance
Genetic variations can affect the immune response to infectious conditions. Recent evidence shows DNA copy number variations (CNVs) in the DEFA1A3 locus, which encodes for human neutrophil peptides 1-3 (HNP1-3) are associated with urinary tract infections (UTIs). A manipulatable human DEFA4/4 gene transgenic mouse model allows further characterization of UTI and pyelonephritis susceptibility. In this study, we demonstrate the expression of DEFA1A3 in the transgenic human DEFA4/4 mouse mirrors human urinary tract DEFA1A3 expression. We also demonstrate this transgenic mouse is protected from uropathogenic Escherichia coli (UPEC) challenges. Additionally, in vitro assays using HNP1-3 against UPEC elucidate combinational effects when used with other antimicrobial peptides. Our models allow for future studies in precision medicine and novel therapeutic development.
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.
Sign up for PNAS alerts.
Get alerts for new articles, or get an alert when an article is cited.
Urinary tract infection (UTI) occurs in 50 to 60% of women worldwide and susceptibility factors are associated with heritable genetic alterations in the innate immune defenses, including antimicrobial peptides (AMPs), pattern recognition receptors, inflammatory mediators, and phagocytes (1). The alpha-defensins 1, 2, and 3 DEFA1A3 locus encodes human neutrophil peptide 1-3 (HNP1-3). This pleiotropic AMP exerts potent microbicidal, chemotactic, and immunomodulatory activities in both in vitro and in vivo models of infection and inflammation (2–5). Recent evidence supports that DNA copy number variations in the DEFA1A3 locus are associated with UTI susceptibility in children with vesicoureteral reflux (VUR) (6). Because alpha-defensins 1-3 are expressed in both myeloid and kidney epithelia cell sources, the relative importance of each contributor in the host defense response against UTIs is largely unknown. The absence of a murine DEFA1A3 gene homolog has resulted in limited knowledge regarding the biological relevance under the immune response against uropathogens. In this report, we utilize a human gene transgenic mouse that expresses the human DEFA1A3 gene under uropathogenic Escherichia coli model challenges to elucidate its role in UTI defense from invasion and colonization of the urinary tract. We further support our findings by elucidating synergistic effects of HNP1-3 with the human antimicrobial peptides cathelicidin (LL-37) and ribonuclease7 (RNAse7) when tested against various UPEC isolates under in vitro conditions. Collectively, our in vivo multidrug resistance murine challenges support the synergistic activity of DEFA1A3 combining direct antimicrobial and immunomodulatory AMP activity to protect the murine UT. We postulate several possible mechanisms in which UPEC burden and attachment could be acting upon in vivo and under the presence of renal-derived HNP1-3 production.
Methods
Experiments, Figure Design, and Statistical Analyses.
Experimental UTI challenges were performed by transurethrally injecting an inoculum of 50 μL into the bladder of female 6- to 12-wk-old mice. Alternatively, the equivalent volume of sterile phosphate-buffered saline (PBS) was administered transurethrally to control mice. For double inoculum experiments, repeat infections were performed 3 h following the first administration. Mice were killed by CO2 inhalation and bilateral nephrectomy for organ harvesting and processing. Further information and references on methodologies and materials used in the experiments performed can be found in SI Appendix. Figures were plotted using GraphPad Prism 8 (GraphPad Software). *P value <0.05, **P value <0.01, ***P value <0.001, and ****P value <0.0001 were considered statistically significant. Statistical significances between groups were determined by software’s two-tailed Student’s t test unless specified in the figure legend or methods section. The schematic art images used in figures were obtained from Servier Medical art (https://servier.com/Powerpoint-image-bank). Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License.
Results
Whole-Genome Sequence Analysis Localizes the DEFA1A3 Locus to the Single Insertion Site on Chromosome 13 of the Transgenic Human DEFA4/4 Gene Mice.
We performed whole-genome sequencing of genomic DNA from both wild-type (WT) and transgenic human DEFA4/4 gene mice using long-range sequencing technology (10× Genomics). The sequence alignment analysis shows that DEFA1A3-containing reads map to chromosome 13 of the mouse genome. We then aligned the flanking sequences of these reads onto the University of California Santa Cruz genome assembly of the mouse (GRCm38/mm10). Analysis determined a single insertion site of the DEFA1A3 cassette to be located within the second intron of the mouse gene VMn1r195 on chromosome 13 (Fig. 1A). The copy number (CN) of genes was estimated to be eight CNs for the insertion cassette localized in the DEFA4/4 genome on the C57BL/6J strain mouse background. To confirm our alignment sequencing results, we performed polymerase chain reaction (PCR) genotyping using a primer-probe set designed out of the flanking sequences of the insertion sites for the target repeats. Agarose gel products resulted in 729-bp amplicon bands for nonrepeat carriers. A band of 513 bp was reproduced to determine repeat carriers in the transgenic human DEFA4/4 gene mouse (Fig. 1 B and C).
Fig. 1.
Baseline Inflammatory, Antimicrobial Peptide Expression Markers, Immune Cell Profile, and Gastrointestinal Microbiota Composition Are Not Influenced by Insertion of the Human DEFA1A3 Locus.
We evaluated the effects of transgene insertion in the transgenic human DEFA4/4 gene mouse compared to its WT counterpart utilizing inflammatory and AMP expression using mRNA markers of the target urinary tract. We further support these findings by performing gastrointestinal (GI) microbiota composition using 16S sequencing alignment by phylum, and immune cell profile using myeloid and nonmyeloid surface markers expressed in kidneys. We found no statistically significant differences in the inflammatory profiling of the kidney and bladder compared to WT or in the GI microbiota amplicons sequenced (SI Appendix, Figs. S1 and S2).
Experimentally Induced Pyelonephritis Increases DEFA1A3 mRNA Expression in the Kidney and Bladder of Infected Transgenic Human DEFA4/4 Gene Mouse.
We examined the role of the DEFA1A3 gene locus in the transgenic human DEFA4/4 gene mouse by modeling UTI responses in the human kidney upon UPEC challenge with the CFT073 pyelonephritis strain. We conducted time-point studies to evaluate the regulation of alpha-defensins transcription kinetics in the transgenic human DEFA4/4 gene mouse kidney and bladder tissues. DEFA1A3 mRNA transcripts significantly increased in the kidney and bladder by 1.98 ± 0.16-fold increase (P < 0.0001) and 10.47 ± 2.83-fold increase (P < 0.0001), respectively, compared to unstimulated controls (Fig. 2 A and B). The kidney peak transcription occurred 6 h postinfection (hpi), while bladder expression peaked at 24 hpi. Interestingly, down-regulation of DEFA1A3 transcription in kidney and bladder occurred at multiple early time points. Transcriptional regulation of kidney and bladder tissues of transgenic human DEFA4/4 gene mouse were restored to baseline levels by 48 h following UPEC pyelonephritis challenge. We also evaluated other peptides important to innate immunity that are induced in the kidney following UPEC challenge (7). We compared the induction of pentraxin 3 (Ptx3), lipocalin 2 (Lcn2), myeloperoxidase (Mpo), and β-defensins 1 (Defb) mRNA AMPs upon experimental pyelonephritis challenge at the acute time points between the DEFA4/4 and C57BL/6J mouse kidneys (SI Appendix, Fig. S3). Interestingly, Mpo and Defb mRNA expression was induced as early as 1 and 3 hpi, respectively. Significant induction of Lcn2 and Ptx3 occurred at 6 hpi and returned to baseline levels after 24 h of challenge. Moreover, mean expression levels were not statistically different between WT and DEFA4/4-infected kidneys for the AMP mRNA markers assessed. These findings indicate that DEFA1A3 does not induce other components of the innate immune system involved in the defense against UTIs. To determine whether our model recapitulates HNP1-3 secretion into urine during infection as seen in UTI patients, we designed a sandwich enzyme-linked immunosorbent assay (ELISA) with urine samples from infected transgenic DEFA4/4 mice (6). Due to limitations in mouse clean-catch urine collection methods, samples of four mice per group were pooled to quantify urinary HNP1-3 levels. Urine samples from DEFA4/4 at 24 h post-UPEC challenge showed elevation of HNP1-3 compared to saline-injected controls regardless of the UPEC strains inoculated (P = 0.341). Urine HNP1-3 levels significantly increased for both UTI89 (P = 0.004) and CFT073 (P = 0.001) experimental UTI challenges when compared to saline controls (Fig. 2C). Immunofluorescence experiments visually showed enhanced expression of HNP1-3 mostly colocalized in aquaporin-2 (AQP2) positive collecting ducts in kidney sections from transgenic human DEFA4/4 gene mouse upon UPEC pyelonephritis challenge at 6 hpi compared to baseline expression in saline-injected mice (Fig. 2D). At this time point, we also observed other nonstructural HNP1-3–expressing cells upon infection, irrespective of myeloid cell migration (Fig. 2E). The expression of HNP1-3 and AQP2 dual-positive cells appears in apical collecting duct cells postpyelonephritis challenge in DEFA4/4 mice compared to saline controls. Additionally, we evaluated immunohistochemistry staining of transgenic human DEFA4/4 gene mouse bladders. Although we did not detect baseline HNP1-3 expression in epithelial cells, there was a significant influx of HNP1-3–expressing nonstructural cells visually localized around the bladder lumen (Fig. 2 F and G). To further understand the contribution of different cellular sources of alpha-defensins under the UTI setting, we performed in vivo experimental UTI challenges of our transgenic mice and evaluated mRNA expression levels of UPEC groups of three cellular subsets (CD45+, CD45−cKIT+, and CD45−cKIT−). Interestingly, the myeloid (CD45+) and intercalated cell populations (CD45−cKIT+) had the highest DEFA1A3 transcription at baseline. The myeloid cell populations (CD45+) had no change in transcriptional expression of DEFA1A3 under the pyelonephritis UTI murine challenge at 6 hpi (P = 0.99), whereas the intercalated cell population increased by 14.17-fold ± 14.81 (P = 0.33) when compared to saline transgenic controls (SI Appendix, Fig. S4A). We further confirmed this transcription pattern in enriched human intercalated cells using similar cell population sorting methodologies (8) and identified a significant up-regulation of DEFA1A3 mRNA with a mean increase of 2.93-fold ± 0.62 (P = 0.02) following in vitro stimulation with UPEC; the CFT073 strain compared to cells under saline treatment derived from four separate human kidney tissue samples (SI Appendix, Fig. S4B).
Fig. 2.
Transgenic Human DEFA4/4 Gene Murine Urinary Tract Demonstrates Enhanced Protection against UPEC Kidney and Bladder Infection.
We quantified and compared the bacterial burden of kidneys and bladders for the infected transgenic human DEFA4/4 gene and C57BL/6J mice (WT). Experiments were conducted on transurethrally infected mice with two UPEC strains for selective infectivity toward the kidney (CFT073) or bladder (UTI89). To evaluate early invasion and persistence of UPEC in urinary tract tissues, we evaluated colony forming unit (CFU) per gram of bladders and left and right kidneys at 6 and 24 hpi between mouse genotypes. First, transgenic human DEFA4/4 gene mice transurethrally challenged with cystitis strain UTI89 demonstrated decreased bladder and kidney burdens at 6 h. The kidney bacterial clearance was higher in DEFA4/4 mice compared to WT (Fig. 3 A and B). In the challenge with the more virulent pyelonephritis strain CFT073, the protection of DEFA4/4 mice kidney and bladder tissues was more evident when compared to the WT mouse. At early time points, both the kidney and bladder demonstrate lower burdens, but only the mean bladder CFU burdens were significantly lower in the DEFA4/4 mouse. At 24 h, the bladders had significantly lower bacterial burdens compared to WT controls (Fig. 3 C and D). Similar to the UTI89 UPEC cystitis strain challenge, the CFT073 UPEC pyelonephritis strain challenge showed no significant differences at later time points evaluated 1 wk postinfection (SI Appendix, Fig. S5 A and B). After 1 wk postinfection, we did not observe differences in signs of renal scarring between infected transgenic and WT mice in kidney Masson’s trichrome stained slides (SI Appendix, Fig. S5 C–E). Due to limited renal infection burden in the human gene transgenic mouse model of UTIs compared to WT, we further induced experimental pyelonephritis twice by repeating the UTI challenge in the transgenic mice 3 h following the first challenge and were able to find an inverse correlation between peak kidney DEFA1A3 mRNA expression at 6 hpi with CFU counts for the bladder (r2 = 0.545) and kidney (r2 = 0.608) tissue lysates, respectively (Fig. 3 E and F). Additionally, we evaluated the gene-dosage effect of the DEFA1A3 genome copy number (GCN) insertions under the pyelonephritis UTI murine challenge at 6 hpi using littermate homozygous DEFA4/4, heterozygote DEFA4/0, and wild-type genotypes (8, 4, and 0 GCNs per diploid genome, respectively). We found significantly lower bacterial burden and bacterial clearance in the hemizygous DEFA4/0 mouse group with four GCNs when compared to littermate zero human gene transgenic mouse urinary tract tissues lacking DEFA1A3 expression, respectively, for bladder (P = 0.0004) and kidneys (P = 0.022) at 6 hpi. No significant differences were found between eight and four GCN groups in mean bacterial burden of bladder (P = 0.20) or kidneys (P = 0.81), although more kidneys were protected from infection in the eight GCN (DEFA4/4) when compared to four GCN (DEFA4/0) group (Fig. 3 G and H). In order to determine whether renal sources of HNP1-3 alone are enough to protect the urinary tract, we performed kidney transplant isographs into WT recipient mice with DEFA4/4 transgenic kidneys and used WT transplants to WT recipients as controls for infection studies (Fig. 3I). Following experimental pyelonephritis challenge, the DEFA4/4 transgenic kidney transplanted WT recipient mice group exerted significant reduction in mean bladder total CFU burden (P = 0.002), and kidney bacterial protection in the transgenic kidney transplanted mice when compared to control transplants at 6 hpi (Fig. 3 J and K). The resistance of a WT kidney into a WT mouse to ascending UPEC infection using our methodology is consistent with our prior studies using a transplant infection model (9).
Fig. 3.
DEFA1A3 Does Not Magnify Kidney Inflammation following UPEC Challenge.
From the single and double inoculation experimental pyelonephritis challenges in DEFA4/4 and WT mice, we examined kidney histology in order to measure the extent of pyelonephritis in infected kidneys. Kidney sections stained with hematoxylin and eosin dyes (H&E) were evaluated by implementing a previously reported inflammatory scoring system (10, 11). We blindly scored tissue sections assessing the medullary and cortex kidney regions from each kidney from respective genotypes (SI Appendix, Fig. S6 A–H). We did not find inflammatory differences between infected DEFA4/4 and WT mice at 6 or 24 hpi (SI Appendix, Fig. S6I). At these acute time points, inflammation was most prominent in the kidneys from mice with double inoculations regardless of the mouse genotypes at 6 and 24 hpi. Then, we correlated inflammatory scores, kidney CFU burdens, and relative mRNA expression of interleukin 6 (Il6), C-C motif ligand 2 (Ccl2), interleukin 1-beta (Il1β), and tumor necrosis factor alpha (Tnfα) mRNA inflammatory markers in C57BL6/J kidneys. We are able to demonstrate significant correlations between murine inflammatory kidney scores, CFU bacterial titers, Il6, and Il1β mRNA inflammatory markers following single and double CFT073, UPEC challenges as early as 6 hpi (SI Appendix, Table 1). Correlated in C57BL6/J mice, inflammatory scores correlated the most with Il6 and Il1β mRNA fold expression (R2 = 0.728, P = 0.003 and R2 = 0.828, P = 0.0007, respectively). Additionally, Il6 and Il1β strongly correlated to kidney CFU bacterial burdens (R2 = 0.881, P = 0.0002 and R2 = 0.602, P = 0.014). Using these inflammatory assessment methodologies, enabled us to correlate infection and inflammation with the relative expression of Il6 and Il1β cytokines.
We then compared the correlations between inoculated C57BL6/J and DEFA4/4 kidneys infected at 6 hpi with the pyelonephritis CFT073 strain. Following 6 hpi, kidneys from infected DEFA4/4 mice wild-type mice had statisitically significant correlation between kidney Il6 and Il1β cytokine mRNA expression and CFU bacterial burdens (SI Appendix, Table 2). Additionally, we were able to demonstrate that following 6 hpi with CFT073 infection, the DEFA4/4 kidneys exerted significantly lower expression of Il6 and Il1β mRNA inflammatory markers upon infection mice compared to wild-type kidneys (P = 0.0437 and P = 0.0092 for each cytokine) (SI Appendix, Fig. S7 A and B). Cumulatively, these results suggest that the HNP1-3 AMP has direct protection from UPEC challenge and not from a secondary effect of inducing inflammatory pathways.
Human Neutrophil Peptides 1-3 Exert Potent In Vitro Antimicrobial Activity by Inhibiting Uropathogenic E. coli Growth, Attachment, and Direct Killing.
We utilized natural HNP1-3, a commercially available peptide, to determine the minimum inhibitory concentration breakpoints against enteropathogenic ATCC10789/K-12 (EC), and UPEC UTI89 and CFT073 E. coli strains under Clinical and Laboratory Standards Institute (CLSI) guidelines (SI Appendix, Fig. S8 A and B). We identified strain-selective resistance against HNP1-3 reflective of other antimicrobial peptides (Fig. 4A). To better recapitulate the urinary microenvironment, we adjusted the sodium concentration, level of acidity, and alkalinity of the growth media. As a result, raising the pH of media to 9.0 resulted in an average of 30 to 40% antimicrobial activity regardless of the isolate tested at the respective minimal inhibitory concentration (MIC), which represents the smallest concentration of a substance to stop bacterial growth (SI Appendix, Fig. S8D). We report no change in antimicrobial activity when pH was lowered to 4.0. Additionally, addition of sodium chloride at 150 mM in the growth media reduced antimicrobial activity by an average of 50 to 60% (SI Appendix, Fig. S8E). To assess whether HNP1-3 protects the urothelium from bacterial attachment, confluent human bladder epithelial cells (HBLAK) were challenged with UPEC in the presence and absence of HNP1-3. Compared with the vehicle control, the percent of UPEC adhered to HBLAK cells incubated with HNP-1 was significantly reduced at 2× MIC but not at 1× MIC (P = 0.650) (Fig. 4B). To confirm whether HNP1-3 kills UPEC in vitro, we collected cell culture media inoculated with UPEC in the presence and absence of HNP1-3. After a 2-h incubation with HNP1-3, UPEC survival was significantly decreased with both 1× and 2× MIC (Fig. 4C).
Fig. 4.
Human Neutrophil Peptides 1-3 Are Synergistic with Cathelicidin In Vitro against Uropathogenic and Multidrug Resistant E. coli Isolates.
Following determination of MIC breakpoints with broth macrodilution and microdilution assays for LL-37, RNAse7, beta-defensins 1 and beta-defensins 2 against E. coli isolates, combinations of antimicrobial peptides were tested at twofold increases to determine synergistic (<0.5), additive (0.5 to 1.0), or no interaction between AMPs and HNP1-3 (SI Appendix, Fig. S8C). Regardless of the E. coli isolate, the calculated fractional inhibitory concentration indexes (FICIs) indicated synergistic effects of LL-37 following the addition of sub-MIC of HNP1-3 (FICI = 0.37) with the exception of MDR58 (FICI = 0.5), which was additive. Combinations of sub-MICs with RNAse7 demonstrated additive effects among all strains (FICI = 0.5). Interestingly, there was no interaction when adding human beta-defensin 1 or human beta-defensin 2 (FICI = 1.0) (Fig. 4 D–G). Evidenced from the drug resistance phenotype from our in vitro MIC breakpoint studies using the MDR58 strain, we tested in vivo antimicrobial resistance in our pyelonephritis model by inoculating MDR58 into DEFA4/4 and C57BL6/J mice (Fig. 4 H and I). At 6 hpi, quantified bladder and individual kidney bacterial burdens showed a nonsignificant decrease of mean bacterial colonization in the DEFA4/4 compared to the wild-type counterpart in both kidneys and bladders (mean = 5.26 ± 0.843 and 5.582 ± 0.582 for bladders and mean = 1.44 ± 2.070 and 2.31 ± 2.561 for kidneys). Additionally, we correlated kidney CFU and previously mentioned inflammatory mRNA markers (SI Appendix, Table S3). Our results indicate that inflammatory mRNA expression in wild-type mice is higher than in transgenic DEFA4/4 infected with MDR58-infected kidneys, in which induction of Il1β and Il6 gene expression was consistent with mice infected with the CFT073 strain (SI Appendix, Fig. S7 E–H). Taken together, our results support that DEFA1A3 protects the murine urinary tract by combining antimicrobial and potential downstream immunomodulatory effects.
Discussion
In this study, we utilized long-range genomic sequencing to localize a single insertion site of the DEFA1A3 locus into the C57BL/6J genome with eight genome copy numbers in the insertion cassette. We also demonstrate that this human gene transgenic mouse mimics the transcriptional DEFA1A3 messenger RNA and alpha-defensins 1-3 antimicrobial peptide up-regulation as presented in humans with UTIs. Additionally, we confirmed that the renal sources of human HNP1-3 in human gene transgenic DEFA4/4 mice can protect the urinary tract from UPEC challenge at early infection time points in an orthotopic kidney transplant model. We showed direct antimicrobial properties against uropathogenic E. coli in host cell attachment and effects of combining HNP1-3 with other AMPs found to be expressed in renal sources under UTI settings (12, 13), primarily associating strong synergy interactions with minimum subinhibitory concentrations of cathelicidin (LL-37) peptide against various E. coli isolates under in vitro conditions. Finally, the human gene transgenic DEFA4/4 mice lower urinary tracts were not significantly protected when challenged with multidrug resistant UPEC MDR58, expanding the current understanding of in vivo AMP bacterial resistance in a murine model of UTIs. Collectively, these findings show the potent antimicrobial activity coupled with immunomodulatory functions of α-defensins 1-3 expression protects the kidney upon infection. These findings allow for the further expansion of precision medicine and therapeutic development opportunities focusing on the association of the DEFA1A3 locus and its role as potent antimicrobial peptide in UTI protection of the urinary tract.
Here, we provide the informatic and sequencing pipelines for mixed species human gene transgenic models. By using alignment for sequenced reads, we confirm the target insertion sites of the DEFA1A3 cassette into the mouse genome at chromosome 13. This approach can be recapitulated in other human gene transgenic murine models. We then generated primer-probe sets at flanking regions of insertion sites for the genotyping of hemizygous and noncarrier transgenic mice through PCR methodologies. Additionally, we provide comparable marker levels for expression of inflammatory or antimicrobial peptide effectors and immune cell populations in the target urinary tract tissues between the human gene transgenic mice and the wild-type counterpart. We further assessed the histology of infected kidneys with a scoring system that correlated with inflammatory mRNA markers (Il6 and Il1β) during acute time points. We found no measurable differences in the histologic assessment of infected kidneys between mouse genotypes. Using the coupled assessment, we were able to better correlate the bacterial loads in the infected kidney with mRNA cytokines involved in immune cell recruitment. In mice, Il6 and Il1β cytokines have been associated with the activation of the intracellular NLRP3 inflammasome to promote renal inflammation and damage (14). Our results indicate the consistent down-regulation of proinflammatory Il1β signaling. We speculate different levels of Il6 expression can be from its pleotropic pro- and antiinflammatory effects during UTI to protect the kidney from increased bacterial burden (15). Future studies will involve exploration of different immune cell types involved in the immunomodulation properties of DEFA1A3 against UPEC at various in vivo time points. Furthermore, urinary IL6 and IL1β have been reported as biomarkers for patients with recurrent and drug-resistant UTIs (15–17). Our assessment of inflammation and mRNA cytokine correlations under the transurethral UTI challenge model further supports the induction of markers to enable the early detection of UPEC-associated induction of kidney inflammation in mice. Our findings using the MDR58 pyelonephritis strain in DEFA4/4 mice indicate alpha-defensins 1-3 can exert immunomodulatory properties regardless of its exerted antimicrobial activity, which is consistent with other studies suggesting the release of DEFA1A3/HNP1-3 can be an attenuator of inflammation and tissue destructive responses (18, 19) Additionally, DEFA4/4 mice exhibited a decrease of proinflammatory mRNA expression upon acute pyelonephritis compared to control conditions, which should be further explored under various infectious disease models such as ulcerative colitis and sepsis.
Studies have investigated the segregation of low and high DNA copy number variations of the alpha- and beta-defensin genes correlated and associated with susceptibility toward infections in a variety of disease settings (2, 20–24). Our previous study described kidney expression of DEFA1A3 in humans (6). Additionally, we demonstrated that higher DEFA1A3 DNA copy numbers were associated with UTI protection in children. In this study, we demonstrate that DEFA1A3 expression in the transgenic human DEFA4/4 gene mouse mirrors the expression in humans. Our immunofluorescence colocalization results show similar expression patterns along other pyelonephritis studies and compared to previous human studies characterizing the expression of alpha-defensins 1-3 and alpha-defensin 5 in collecting duct cells (6, 25, 26). We also demonstrate that human DEFA1A3 is protective against UPEC invasion in mice, gene-dosage effects associated with genome copy number, and the importance of the renal sources of human HNP1-3 in the defense of urinary tract infection.
We evaluated the use of three UPEC strains in the UTI human gene transgenic mouse model with the aim to identify urinary tract organ-specific responses. We selected UPEC because of two tropism characteristics expressed in the urinary tract are selective toward the bladder, causing cystitis, or ascending through the ureters, resulting in pyelonephritis (27–30). The two strains we used have distinct virulence patterns and were isolated from two different types of patients with UTIs. In our UPEC challenge studies, the bladder infection persisted, and clearance did not differ between DEFA4/4 and WT mice in both early and late time points with UPEC cystitis strain UTI89. This suggests that UTI89 possibly subverts the action of HNP1-3 by an unknown mechanism. We hypothesized that the virulence factors of UTI89 allowed for invasion and sustained infection of the bladder due to increase in vitro resistance to HNP1-3 when comparing uropathogenic and multidrug resistant E. coli isolates. With UPEC pyelonephritis strain CFT073, we observed reduced burden in the DEFA4/4 mouse bladder and kidneys compared to WT. When the MDR58 pyelonephritis strain was inoculated, we did not demonstrate significantly reduced burdens. This finding suggests that CFT073 is susceptible to the antimicrobial action of HNP1-3 at the level of the bladder. The lower burden in the kidneys then could be from a reduced bacteria burden during a cystitis/lower tract UTI resulting in a smaller pool of bacteria to ascend to the kidney and/or a distinct protective effect at the level of the kidney. Therefore, our studies determined the most protective effects of HNP1-3 were against the CFT073 strain, which is interestingly the most virulent. We conducted subsequent UTI challenges for the rest of our murine experiments with CFT073 to better understand the role of HNP1-3 in defense of the kidney. Additionally, we challenged littermate mice containing high (eight), medium (four), or zero human gene transgene copies of the human DEFA1A3 locus and were able to find similarities between bacterial decrease and enhanced clearance medium and high GCN groups at an early time point, elucidating the role of gene copy number variations in the setting of UTI. We utilized our kidney transplant model to model kidney isolated effects during UTI (9). We found that transgenic DEFA4/4 kidney in WT recipients (exclusive kidney expression of HNP1-3) showed consistent resistance to infection from CFT073 specifically with a significant decrease in bladder burdens due to the direct antimicrobial effects of kidney-derived HNP1-3, given that the mice had WT neutrophils. Further studies are needed to better dissect cellular mechanisms driving UTI resolution and whether myeloid sources of HNP1-3 play a role under UTI acute disease phases.
At the level of transcription, we found interesting tissue-specific temporal patterns. Initially 1 h following UPEC challenge, DEFA1A3 transcripts were down-regulated, but by 6 hpi, we saw peak kidney expression levels similar to the induction of beta-defensins in mice challenged with uropathogens (31, 32). We hypothesize that HNP1-3 from alpha-granules of activated resident neutrophils may down-regulate kidney epithelial DEFA1A3 transcription during early infection. In other organ systems, HNP1-3 diminishes macrophage transcriptional machinery, thereby preventing an excessive acute inflammatory response and allowing for more effective phagocytosis of invading pathogens (19, 33, 34). We assessed the mRNA expression of renal AMPs that could possibly be the predominant early response to UTI; however, their transcriptional kinetics were similar between mice genotypes. Although we did not detect gene expression differences in the kidney and bladder for Ptx3, Mpo, Lcn2, and Defb1, we cannot rule out the possibility that alternative AMPs or innate immune effectors could interact with HNP1-3 at early time points. Blocking or knocking out the expression of these kidney-derived AMP candidate genes such as Pentraxin 3 would help to define their contribution to the UTI innate immune responses.
A limitation of using a transurethral inoculation model is the variable incidence of kidney bacterial burden and urine reflux due to the protected phenotype transgenic mouse, which is on a nonrefluxing C57BL/6 background (35). In future studies, models and methodologies will be employed that will allow for elucidation of myeloid versus epithelial DEFA1A3 transcription and translation during UTI (36, 37). Chimeric and kidney allograft transplant models coupled with RNA sequencing (RNA-seq) and proteomic approaches will further define the transcription and relative contributions of alpha-defensins 1-3 from kidney and extrakidney sources upon UPEC challenge.
We tested the antimicrobial potency of natural HNP1-3 against uropathogenic isolates, including multidrug resistant E. coli, although these concentrations surpass the physiologically relevant up-regulation in urine samples. By determination of minimum inhibitory concentration breakpoints and fractional inhibitory concentration index determination using CLSI in vitro methodologies, we were able to elucidate additive and synergistic effects of HNP1-3 with other human antimicrobial peptides. Cathelicidin (LL-37), an AMP expressed and up-regulated under UTI acute phases in human and mouse tissues, had synergistic effects with HNP1-3. Additionally, we were able to establish an additive relationship with ribonuclease 7 (RNase7) peptide, but no interaction with recombinant human beta-defensin 1 or human beta-defensin 2. Further studies are needed to understand and confirm these interactions between inducible and ubiquitously expressed AMPs under the UTI setting in humans. These findings represent potential therapeutic and precision medicine efforts in the future. The kidney and extrakidney sources of HNP1-3 may interact with other antimicrobial peptides released into the urinary tract during UTI to provide enhanced barrier protection (SI Appendix, Fig. S9). Because of the importance of DEFA1A3 in the kidney innate immune response, further research needs to determine whether DEFA1A3 has any cytotoxicity during health, disease, malignancy, infection, or the diverse range of physiologic conditions of the urinary tract. In prior studies DEFA1A3 did not demonstrate cytotoxicity to CD4+ cells in concentrations up to 10 μg/mL, but has been demonstrated cytotoxic to squamous cell carcinoma cells and lung cells in patients with moderate to severe lung disease, including alpha-1 antitrypsin deficiency (38–40). DEFA1A3 cytotoxicity can be a complex issue; for example, HNP1 and LL37/cathelicidin cooperativity has been demonstrated to have the potential to switch from membrane-destructive to membrane-protective functions depending on whether the cell is an enemy or needed host cell (10). We did show that the addition of DEFA1A3 does not increase inflammation at baseline or disrupt the resident immune cell population of the kidney.
Our model has some distinctions from the human condition, and the murine experimental UTI model differs from human UTI in several ways. First, the murine model of UPEC involves a large inoculum delivered instead of natural invasion and propagation of the pathogen growing in vivo over several days and represents an acute early phase of the UPEC infection. Second, protein expression in the transgenic DEFA4/4 mouse is somewhat diminished (70% of total human expression) compared to the human translated forms of alpha-defensins 1-3 (41, 42). Third, quantification of specific intercalated cell subsets in the human gene transgenic mouse collecting duct might be driving the kidney HNP1-3 expression source. Further studies analyzing early expression of urine and blood expression of novel AMPs such as cathelicidins and ribonucleases in the proposed murine models would enable the discovery of potential in vivo interactions with the antimicrobial activity of HNP1-3 upon UPEC colonization of the urinary tract tissues. Because human DEFA1A3 CNVs can range to more than eight copies per genome in some individuals, future studies comparing mouse transgenic genotypes with more than eight GCNs are warranted to expand the murine transgenic model of UTIs and recapitulate translational findings. A major limitation of our study design involves the acknowledgment of a sustained effect from HNP1-3 to suppress UTI chronically as we only measured the significant reduction of bacterial burden in the 6- and 24-hpi acute time points. Studies involving repeated infections and longer time points would provide additional mechanisms of action, which DEFA1A3 might be related to. Lastly, the combinational effects of AMPs and HNP1-3 from our checkerboard studies needs to be investigated under in vivo models.
In summary, the studies reported here demonstrate the protective effects of alpha-defensins gene locus insertion in the transgenic DEFA4/4 mouse against experimentally induced UTI challenge. The kidney parenchyma comprises a dynamic environment in which both resident and infiltrating neutrophils and collecting duct cells may be sources of HNP1-3, generating a powerful and diverse AMP barrier that may neutralize in combination with other AMPs exerting bacterial protection toward uropathogenic invasion. Future studies will be needed to delineate the most biologically relevant source in innate defenses that may provide a novel therapeutic target to up-regulate local expression and model AMP combination effects with commonly prescribed antibiotics to potentially address more effective and safe treatment of UTIs as well as recurrent infections.
Data, Materials, and Software Availability
All study data are included in the article and/or supporting information. All data and protocols are available to interested parties upon request to either corresponding author.
Acknowledgments
This study was supported by National Institute of Diabetes and Digestive and Kidney Diseases 5-R01-DK117934 and 5-R01-DK106286 grants.
Supporting Information
Appendix 01 (PDF)
- Download
- 2.40 MB
References
1
A. H. Qureshi et al., DCHS1 DNA copy number loss associated with pediatric urinary tract infection risk. Innate Immun. 26, 473–481 (2020).
2
Q. Chen et al., Increased gene copy number of DEFA1/DEFA3 worsens sepsis by inducing endothelial pyroptosis. Proc. Natl. Acad. Sci. USA 116, 3161–3170 (2019).
3
T. Hisamatsu et al., Diagnosis and management of intestinal Behçet’s disease. Clin. J. Gastroenterol. 7,205–212 (2014).
4
C. Jespersgaard et al., Alpha-defensin DEFA1A3 gene copy number elevation in Danish Crohn’s disease patients. Dig. Dis. Sci. 56, 3517–24 (2011).
5
N. Mookherjee et al., Antimicrobial host defence peptides: functions and clinical potential. Nat. Rev. Drug Discov. 19, 311–332 (2020).
6
A. L. Schwaderer et al., Polymorphisms in α-defensin-encoding DEFA1A3 associate with urinary tract infection risk in children with vesicoureteral reflux. J. Am. Soc. Nephrol. 27, 3175–3186 (2016).
7
B. Becknell et al., Amplifying renal immunity: the role of antimicrobial peptides in pyelonephritis. Nat. Rev. Nephrol. 11, 642–655 (2015).
8
V. Saxena, et al., Kidney intercalated cells are phagocytic and acidify internalized uropathogenic Escherichia coli. Nat. Commun. 12, 1, 2405 (2021).
9
J. Ketz et al., Developmental loss, but not pharmacological suppression, of renal carbonic anhydrase 2 results in pyelonephritis susceptibility. Am. J. Physiol. Renal Physiol. 318, 1441–1443 (2020).
10
E. Drab, K. Sugihara, Cooperative Function of LL-37 and HNP1 Protects Mammalian Cell Membranes from Lysis. Biophys. J. 119, 2440–2450 (2020).
11
S. E. Bowen, C. L. Watt, I. J. Murawski, I. R. Gupta, S. N. Abraham, Interplay between vesicoureteric reflux and kidney infection in the development of reflux nephropathy in mice. Dis. Model. Mech. 6, 934–941 (2013).
12
T. Eichler et al., Ribonuclease 7 shields the kidney and bladder from invasive uropathogenic Escherichia coli infection. J. Am. Soc. Nephrol. 30, 1385–1397 (2019).
13
K. L. Nielsen et al., Role of urinary cathelicidin LL-37 and human β-defensin 1 in uncomplicated Escherichia coli urinary tract infections. Infect. Immun. 82, 1572–1578 (2014).
14
A. Vilaysane et al., The NLRP3 inflammasome promotes renal inflammation and contributes to CKD. J. Am. Soc. Nephrol. 21, 1732–1744 (2010).
15
C. B. Ching et al., Interleukin-6/Stat3 signaling has an essential role in the host antimicrobial response to urinary tract infection. Kidney Int. 93, 1320–1329 (2018).
16
P. Kannian, V. Ashwini, S. B. Suchithra, K. B. Sindu, Elevated urinary IL-1β levels in multidrug resistant Escherichia coli and Klebsiella infections. Inflamm. Res. 69, 11–13 (2020).
17
S. Hedges et al., Comparison of urine and serum concentrations of interleukin-6 in women with acute pyelonephritis or asymptomatic bacteriuria. J. Infect. Dis. 166, 653–656 (1992).
18
M. Brook et al., Neutrophil-derived alpha defensins control inflammation by inhibiting macrophage mRNA translation. Proc. Natl. Acad. Sci. U.S.A. 113, 4350–4355 (2016).
19
K. Miles et al., Dying and necrotic neutrophils are anti-inflammatory secondary to the release of alpha-defensins. J. Immunol. 183, 2122–2132 (2009).
20
D. Xu, W. Lu, Defensins: A double-edged sword in host immunity. Front. Immunol. 11, 764 (2020).
21
J. Zhao et al., Low-copy number polymorphism in DEFA1/DEFA3 is associated with susceptibility to hospital-acquired infections in critically ill patients. Mediators Inflamm. 2018, 2152650 (2018).
22
L. R. Machado, B. Ottolini, An evolutionary history of defensins: A role for copy number variation in maximizing host innate and adaptive immune responses. Front. Immunol. 6, 115 (2015).
23
M. Li, X. Yu, Genetic study of immunoglobulin A nephropathy: From research to clinical application. Nephrology (Carlton) 23 (suppl. 4), 26–31 (2018).
24
J. D. Spencer, A. L. Schwaderer, B. Becknell, J. Watson, D. S. Hains, The innate immune response during urinary tract infection and pyelonephritis. Pediatr. Nephrol. 29, 1139–1149 (2014).
25
J. D. Spencer et al., Human alpha defensin 5 expression in the human kidney and urinary tract. PLoS One 7, 2 (2012).
26
M. Higazi et al. Opposing effects of HNP1 (α-defensin-1) on plasma cholesterol and atherogenesis. PloS One 15, 4 (2020).
27
M. S. Haugan et al., Growth rate of Escherichia coli during human urinary tract infection: Implications for antibiotic effect. Antibiotics (Basel, Switzerland) 8, 92 (2019).
28
T. J. Wiles et al., Origins and virulence mechanisms of uropathogenic Escherichia coli. Exp. Mol. Pathol. 85, 11–19 (2008).
29
K. Schaale et al., Strain- and host species-specific inflammasome activation, IL-1β release, and cell death in macrophages infected with uropathogenic Escherichia coli. Mucosal Immunol. 9, 124–136 (2016).
30
B. Li et al., Inflammation drives renal scarring in experimental pyelonephritis. Am. J. Physiol. Renal Physiol. 312, F43–F53 (2017).
31
B. Becknell et al., Expression and antimicrobial function of beta-defensin 1 in the lower urinary tract. PloS One 8, 10(2013).
32
R. Bals et al., Mouse beta-defensin 3 is an inducible antimicrobial peptide expressed in the epithelia of multiple organs. Infect. Immun. 67, 3542–3547 (1999).
33
K. Bdeir et al., Neutrophil alpha-defensins cause lung injury by disrupting the capillary-epithelial barrier. Am. J. Respir. Crit. Care Med. 181, 935–946 (2010).
34
O. Soehnlein et al., Neutrophil primary granule proteins HBP and HNP1-3 boost bacterial phagocytosis by human and murine macrophages. J. Clin. Invest. 118, 3491–3502(2008).
35
I. J. Murawski et al., The C3H/HeJ inbred mouse is a model of vesico-ureteric reflux with a susceptibility locus on chromosome 12. Kidney Int. 78, 269–278 (2010).
36
V. Saxena et al., Whole transcriptome analysis of renal intercalated cells predicts lipopolysaccharide mediated inhibition of retinoid X receptor alpha function. Sci. Rep. 9, 545 (2019).
37
C. Chassin, E. Tourneur, M. Bens, A. Vandewalle, A role for collecting duct epithelial cells in renal antibacterial defences. Cell. Microbiol. 13, 1107–1113 (2011).
38
T. L. Chang et al., Dual role of alpha-defensin-1 in anti-HIV-1 innate immunity. J. Clin. Investig. 115, 765–773(2005).
39
S. T. McKeown et al., The cytotoxic effects of human neutrophil peptide-1 (HNP1) and lactoferrin on oral squamous cell carcinoma (OSCC) in vitro. Oral Oncol. 42, 685–690 (2006).
40
M. Wencker, M. L. Brantly, Cytotoxic concentrations of alpha-defensins in the lungs of individuals with alpha 1-antitrypsin deficiency and moderate to severe lung disease. Cytokine 32, 1–6 (2005).
41
R. Abu-Fanne et al., Neutrophil α-defensins promote thrombosis in vivo by altering fibrin formation, structure, and stability. Blood 133, 481–493 (2019).
42
J. M. Bonmatin et al., Two-dimensional 1H NMR study of recombinant insect defensin A in water: Resonance assignments, secondary structure and global folding. J. Biomol. NMR 2, 235–256 (1992).
Information & Authors
Information
Published in
Classifications
Copyright
Copyright © 2022 the Author(s). Published by PNAS. This article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).
Data, Materials, and Software Availability
All study data are included in the article and/or supporting information. All data and protocols are available to interested parties upon request to either corresponding author.
Submission history
Received: April 19, 2022
Accepted: August 8, 2022
Published online: September 26, 2022
Published in issue: October 4, 2022
Keywords
Acknowledgments
This study was supported by National Institute of Diabetes and Digestive and Kidney Diseases 5-R01-DK117934 and 5-R01-DK106286 grants.
Notes
This article is a PNAS Direct Submission. M.Z. is a guest editor invited by the Editorial Board.
Authors
Competing Interests
The authors declare no competing interest.
Metrics & Citations
Metrics
Citation statements
Altmetrics
Citations
Cite this article
119 (40) e2206515119,
Export the article citation data by selecting a format from the list below and clicking Export.
Cited by
Loading...
View Options
View options
PDF format
Download this article as a PDF file
DOWNLOAD PDFLogin options
Check if you have access through your login credentials or your institution to get full access on this article.
Personal login Institutional LoginRecommend to a librarian
Recommend PNAS to a LibrarianPurchase options
Purchase this article to access the full text.