Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Sci Rep ; 12(1): 2005, 2022 02 07.
Article in English | MEDLINE | ID: mdl-35132157

ABSTRACT

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


Subject(s)
Caspase 1/physiology , Caspases, Initiator/physiology , Epithelial Cells/immunology , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Urinary Tract Infections/genetics , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli/immunology , Caspases, Initiator/metabolism , Cell Line , Epithelial Cells/metabolism , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Inflammation , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophils/metabolism , Phagocytosis , Reactive Oxygen Species/metabolism , Urinary Bladder/cytology
2.
Adv Sci (Weinh) ; 9(6): e2103303, 2022 02.
Article in English | MEDLINE | ID: mdl-35018740

ABSTRACT

Innate lymphoid cells (ILCs) are crucial in orchestrating immunity and maintaining tissue homeostasis in various barrier tissues, but whether ILCs influence immune responses in the urinary tract remains poorly understood. Here, bladder-resident ILCs are comprehensively explored and identified their unique phenotypic and developmental characteristics. Notably, bladder-resident ILCs rapidly respond to uropathogenic Escherichia coli (UPEC) infection. It is found that ILC3 is necessary for early protection against UPEC infection in the bladder. Mechanistically, UPEC infection leads to interleukin (IL)-1ß production in the bladder via a MyD88-dependent pathway, which promotes ILC3 activation. ILC3-expressed IL-17A further recruits neutrophils and controls UPEC infection in the bladder. Together, these results demonstrate a critical role for bladder ILCs in the host defense against UPEC infection.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Immunity, Innate/immunology , Urinary Tract Infections/immunology , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/immunology , Animals , Disease Models, Animal , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Urinary Bladder/immunology
3.
Microbiology (Reading) ; 167(10)2021 10.
Article in English | MEDLINE | ID: mdl-34623231

ABSTRACT

Most uropathogenic Escherichia coli (UPEC) express type-1 fimbriae (T1F), a key virulence factor for urinary tract infection (UTI) in mice. Evidence that conclusively associates this pilus with uropathogenesis in humans has, however, been difficult to obtain. We used an experimental porcine model of cystitis to assess the role of T1F in larger mammals more closely related to humans. Thirty-one pigs were infected with UPEC strain UTI89 or its T1F deficient mutant, UTI89ΔfimH, at inoculum titres of 102 to 108 colony forming units per millilitre. Urine and blood samples were collected and analysed 7 and 14 days post-inoculation, and whole bladders were removed at day 14 and analysed for uroepithelium-associated UPEC. All animals were consistently infected and reached high urine titres independent of inoculum titre. UTI89ΔfimH successfully colonized the bladders of 1/6 pigs compared to 6/6 for the wild-type strain. Intracellular UPEC were detectable in low numbers in whole bladder explants. In conclusion, low doses of UPEC are able to establish robust infections in pigs, similar to what is presumed in humans. T1F are critical for UPEC to surpass initial bottlenecks during infection but may be dispensable once infection is established. While supporting the conclusions from mice studies regarding a general importance of T1F in successfully infecting the host, the porcine UTI models' natural high, more human-like, susceptibility to infection, allowed us to demonstrate a pivotal role of T1F in initial establishment of infection upon a realistic low-inoculum introduction of UPEC in the bladder.


Subject(s)
Cystitis/microbiology , Escherichia coli Infections/microbiology , Fimbriae, Bacterial/metabolism , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/pathogenicity , Virulence Factors/metabolism , Animals , Antibodies, Bacterial/blood , Bacterial Load , Colony Count, Microbial , Disease Models, Animal , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/immunology , Gentamicins/pharmacology , Microbial Viability/drug effects , Mutation , Swine , Urinary Bladder/microbiology , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/immunology , Virulence Factors/genetics
4.
Sci Rep ; 11(1): 19263, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34584119

ABSTRACT

Urinary tract infection frequently caused by E. coli is one of the most common bacterial infections. Increasing antibiotic resistance jeopardizes successful treatment and alternative treatment strategies are therefore mandatory. Metformin, an oral antidiabetic drug, has been shown to activate macrophages in the protection against certain infecting microorganisms. Since epithelial cells often form the first line of defense, we here investigated the effect on uroepithelial cells during E. coli infection. Metformin upregulated the human antimicrobial peptides cathelicidin LL-37 and RNase7 via modulation of the TRPA1 channel and AMPK pathway. Interestingly, metformin stimulation enriched both LL-37 and TRPA1 in lysosomes. In addition, metformin specifically increased nitric oxide and mitochondrial, but not cytosolic ROS. Moreover, metformin also triggered mRNA expression of the proinflammatory cytokines IL1B, CXCL8 and growth factor GDF15 in human uroepithelial cells. The GDF15 peptide stimulated macrophages increased LL-37 expression, with increased bacterial killing. In conclusion, metformin stimulation strengthened the innate immunity of uroepithelial cells inducing enhanced extracellular and intracellular bacterial killing suggesting a favorable role of metformin in the host defense.


Subject(s)
Escherichia coli Infections/drug therapy , Metformin/pharmacology , Urinary Tract Infections/drug therapy , Urothelium/drug effects , Antimicrobial Cationic Peptides/metabolism , Cell Line , Cytokines/metabolism , Drug Repositioning , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Humans , Immunity, Innate/drug effects , Metformin/therapeutic use , Ribonucleases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , TRPA1 Cation Channel/metabolism , Up-Regulation/drug effects , Up-Regulation/immunology , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/immunology , Urothelium/immunology , Urothelium/microbiology , Cathelicidins
5.
Eur J Immunol ; 51(9): 2218-2224, 2021 09.
Article in English | MEDLINE | ID: mdl-34268737

ABSTRACT

Neutrophils play a crucial role in immune defense against and clearance of uropathogenic Escherichia coli (UPEC)-mediated urinary tract infection, the most common bacterial infection in healthy humans. CD300a is an inhibitory receptor that binds phosphatidylserine and phosphatidylethanolamine, presented on the membranes of apoptotic cells. CD300a binding to phosphatidylserine and phosphatidylethanolamine, also known as the "eat me" signal, mediates immune tolerance to dying cells. Here, we demonstrate for the first time that CD300a plays an important role in the neutrophil-mediated immune response to UPEC-induced urinary tract infection. We show that CD300a-deficient neutrophils have impaired phagocytic abilities and despite their increased accumulation at the site of infection, they are unable to reduce bacterial burden in the bladder, which results in significant exacerbation of infection and worse host outcome. Finally, we demonstrate that UPEC's pore forming toxin α-hemolysin induces upregulation of the CD300a ligand on infected bladder epithelial cells, signaling to neutrophils to be cleared.


Subject(s)
Escherichia coli Infections/prevention & control , Neutrophils/immunology , Receptors, Immunologic/deficiency , Receptors, Immunologic/immunology , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli/immunology , Animals , Apoptosis/immunology , Escherichia coli Infections/immunology , Escherichia coli Proteins/metabolism , Female , Gene Knockout Techniques , Hemolysin Proteins/metabolism , Mice , Mice, Inbred BALB C , Phagocytosis/genetics , Phagocytosis/immunology , Phosphatidylethanolamines/metabolism , Phosphatidylserines/metabolism , Receptors, Immunologic/genetics , Urinary Bladder/immunology , Urinary Bladder/microbiology , Urinary Bladder/pathology , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/growth & development
6.
Nat Commun ; 12(1): 2405, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893305

ABSTRACT

Kidney intercalated cells are involved in acid-base homeostasis via vacuolar ATPase expression. Here we report six human intercalated cell subtypes, including hybrid principal-intercalated cells identified from single cell transcriptomics. Phagosome maturation is a biological process that increases in biological pathway analysis rank following exposure to uropathogenic Escherichia coli in two of the intercalated cell subtypes. Real time confocal microscopy visualization of murine renal tubules perfused with green fluorescent protein expressing Escherichia coli or pHrodo Green E. coli BioParticles demonstrates that intercalated cells actively phagocytose bacteria then acidify phagolysosomes. Additionally, intercalated cells have increased vacuolar ATPase expression following in vivo experimental UTI. Taken together, intercalated cells exhibit a transcriptional response conducive to the kidney's defense, engulf bacteria and acidify the internalized bacteria. Intercalated cells represent an epithelial cell with characteristics of professional phagocytes like macrophages.


Subject(s)
Epithelial Cells/immunology , Kidney/immunology , Phagocytes/immunology , Uropathogenic Escherichia coli/immunology , Acids/chemistry , Animals , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Profiling/methods , Humans , Kidney/cytology , Kidney/metabolism , Kidney Tubules, Collecting/cytology , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Phagocytes/cytology , Single-Cell Analysis/methods , Vacuolar Proton-Translocating ATPases/metabolism
7.
Int Immunopharmacol ; 96: 107638, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33848909

ABSTRACT

The majority of urinary tract infections (UTIs) are caused by uropathogenic Escherichia coli (UPEC). Designing a vaccine will certainly reduce the occurrence of infection and antibiotic resistance of the isolates. Antigen 43 (Ag43) and autotransporter H (UpaH) have been associated with the virulence of UPEC. In the present study, the efficacy of different formulations of a hybrid protein composed of Ag43 and UpaH with and without alum and 1,25(OH)2D3 (Vitamin D3) adjuvants were evaluated in mice model. A significant increase in IgG and cellular responses was developed against Ag43::UpaH as compared to the control mice. The addition of alum or a mixture of alum and Vitamin D3 to the protein significantly enhanced the serum IgG responses and tended to remain in a steady state until 6 months. In addition, the mentioned formulations produced significant amounts of IgG1, IL-4, and IL-17 as compared to the fusion protein alone. In addition to the mentioned formulations, the combination of protein with Vitamin D3 also resulted in significantly higher serum IgA and IFN-γ levels as compared to the fusion protein alone. Mice immunized with fusion plus alum and formulation protein admixed with both alum and Vitamin D3 significantly reduced the bacterial load in the bladders and kidneys of mice as compared to the control. In this study, for the first time, the ability of a novel hybrid protein in combination with adjuvants alum and Vitamin D3 was evaluated against UPEC. Our results indicated that fusion Ag43::UpaH admixed with alum and Vitamin D3 could be a promising candidate against UTIs.


Subject(s)
Alum Compounds/pharmacology , Cholecalciferol/pharmacology , Escherichia coli Proteins/immunology , Recombinant Fusion Proteins/immunology , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/immunology , Virulence Factors/immunology , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Alum Compounds/administration & dosage , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/urine , Antigens, Bacterial/immunology , Bacterial Load/drug effects , Bacterial Load/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Bacterial Vaccines/isolation & purification , Cholecalciferol/administration & dosage , Cytokines/metabolism , Immunity, Humoral/drug effects , Immunization/methods , Immunoglobulin G/blood , Immunoglobulin G/urine , Injections, Intravenous , Mice, Inbred BALB C , Mucous Membrane/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/isolation & purification , Urinary Tract Infections/immunology
8.
Front Immunol ; 12: 582858, 2021.
Article in English | MEDLINE | ID: mdl-33679734

ABSTRACT

The structural and functional destruction of the blood-testis barrier (BTB) following uropathogenic E. coli (UPEC) infection may be a critical component of the pathologic progress of orchitis. Recent findings indicate that the mammalian target of the rapamycin (mTOR)-signaling pathway is implicated in the regulation of BTB assembly and restructuring. To explore the mechanisms underlying BTB damage induced by UPEC infection, we analyzed BTB integrity and the involvement of the mTOR-signaling pathway using in vivo and in vitro UPEC-infection models. We initially confirmed that soluble virulent factors secreted from UPEC trigger a stress response in Sertoli cells and disturb adjacent cell junctions via down-regulation of junctional proteins, including occludin, zonula occludens-1 (ZO-1), F-actin, connexin-43 (CX-43), ß-catenin, and N-cadherin. The BTB was ultimately disrupted in UPEC-infected rat testes, and blood samples from UPEC-induced orchitis in these animals were positive for anti-sperm antibodies. Furthermore, we herein also demonstrated that mTOR complex 1 (mTORC1) over-activation and mTORC2 suppression contributed to the disturbance in the balance between BTB "opening" and "closing." More importantly, rapamycin (a specific mTORC1 inhibitor) significantly restored the expression of cell-junction proteins and exerted a protective effect on the BTB during UPEC infection. We further confirmed that short-term treatment with rapamycin did not aggravate spermatogenic degeneration in infected rats. Collectively, this study showed an association between abnormal activation of the mTOR-signaling pathway and BTB impairment during UPEC-induced orchitis, which may provide new insights into a potential treatment strategy for testicular infection.


Subject(s)
Blood-Testis Barrier/immunology , Escherichia coli Infections/immunology , Mechanistic Target of Rapamycin Complex 1/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli/immunology , Animals , Blood-Testis Barrier/metabolism , Cells, Cultured , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Orchitis/immunology , Orchitis/metabolism , Orchitis/microbiology , Rats, Sprague-Dawley , Sertoli Cells/immunology , Sertoli Cells/metabolism , Sertoli Cells/microbiology , Spermatogenesis/immunology , Testis/immunology , Testis/metabolism , Tight Junction Proteins/immunology , Tight Junction Proteins/metabolism , Urinary Tract Infections/metabolism , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/physiology
9.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33653961

ABSTRACT

Given the high frequency of urinary tract infections (UTIs) and their recurrence, there is keen interest in developing effective UTI vaccines. Currently, most vaccine studies, including those in humans, involve parenteral vaccination aimed at evoking and sustaining elevated levels of systemic antibody directed at the uropathogens. In view of recent reports of aberrant Th2-biased bladder immune responses to infection, we hypothesized that immunizing mice intravesically with antigens from uropathogenic Escherichia coli (UPEC) combined with a Th1-skewing adjuvant could correct this defect and promote protection against UTIs. Here we report that compared with mice immunized subcutaneously with this vaccine combination, intravesically immunized mice were markedly more protected from UTIs because of their distinctive ability to recruit Th1 cells into the bladder. This mode of vaccination was effective even in mice that experienced multiple UTIs and displayed pronounced aberrant bladder immune responses. Thus, intravesical vaccination with one or more UPEC antigens to induce bladder Th1 responses represents a superior strategy to combat UTIs, especially in UTI-prone subjects.


Subject(s)
Escherichia coli Infections , Escherichia coli Vaccines/pharmacology , Th1 Cells/immunology , Urinary Bladder/immunology , Urinary Tract Infections , Uropathogenic Escherichia coli/immunology , Animals , Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Escherichia coli Vaccines/immunology , Mice , Mice, Knockout , Urinary Tract Infections/immunology , Urinary Tract Infections/prevention & control
10.
PLoS Pathog ; 17(3): e1009481, 2021 03.
Article in English | MEDLINE | ID: mdl-33788895

ABSTRACT

TcpC is a virulence factor of uropathogenic E. coli (UPEC). It was found that TIR domain of TcpC impedes TLR signaling by direct association with MyD88. It has been a long-standing question whether bacterial pathogens have evolved a mechanism to manipulate MyD88 degradation by ubiquitin-proteasome pathway. Here, we show that TcpC is a MyD88-targeted E3 ubiquitin ligase. Kidney macrophages from mice with pyelonephritis induced by TcpC-secreting UPEC showed significantly decreased MyD88 protein levels. Recombinant TcpC (rTcpC) dose-dependently inhibited protein but not mRNA levels of MyD88 in macrophages. Moreover, rTcpC significantly promoted MyD88 ubiquitination and accumulation in proteasomes in macrophages. Cys12 and Trp106 in TcpC are crucial amino acids in maintaining its E3 activity. Therefore, TcpC blocks TLR signaling pathway by degradation of MyD88 through ubiquitin-proteasome system. Our findings provide not only a novel biochemical mechanism underlying TcpC-medicated immune evasion, but also the first example that bacterial pathogens inhibit MyD88-mediated signaling pathway by virulence factors that function as E3 ubiquitin ligase.


Subject(s)
Escherichia coli Proteins/metabolism , Myeloid Differentiation Factor 88/metabolism , Signal Transduction/physiology , Uropathogenic Escherichia coli/pathogenicity , Virulence Factors/metabolism , Animals , Cell Line , Female , Humans , Immune Evasion/physiology , Macrophages , Mice , Mice, Inbred C57BL , Pyelonephritis/immunology , Pyelonephritis/microbiology , Toll-Like Receptors/metabolism , Ubiquitin-Protein Ligases/metabolism , Uropathogenic Escherichia coli/immunology , Uropathogenic Escherichia coli/metabolism , Virulence/physiology
11.
Sci Rep ; 10(1): 21837, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33318544

ABSTRACT

The NLRP3 inflammasome and IL-1ß have recently been linked to the severity of uropathogenic Escherichia coli (UPEC)-mediated urinary tract infection (UTI). However, not much is known about the contribution of NLRP3 to the antimicrobial properties of neutrophils and the release of IL-1ß during UPEC infection. The purpose of this study was to elucidate the mechanisms behind UPEC-induced IL-1ß release from human neutrophils, and to investigate the contribution of the NLRP3 inflammasome in neutrophil-mediated inhibition of UPEC growth. We found that the UPEC strain CFT073 increased the expression of NLRP3 and increased caspase-1 activation and IL-1ß release from human neutrophils. The IL-1ß release was mediated by the NLRP3 inflammasome and by serine proteases in an NF-κB-and cathepsin B-dependent manner. The UPEC virulence factors α-hemolysin, type-1 fimbriae and p-fimbriae were all shown to contribute to UPEC mediated IL-1ß release from neutrophils. Furthermore, inhibition of caspase-1 and NLRP3 activation increased neutrophil ROS-production, phagocytosis and the ability of neutrophils to suppress UPEC growth. In conclusion, this study demonstrates that UPEC can induce NLRP3 and serine protease-dependent release of IL-1ß from human neutrophils and that NLRP3 and caspase-1 can regulate the antimicrobial activity of human neutrophils against UPEC.


Subject(s)
Interleukin-1beta/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neutrophils/immunology , Phagocytosis , Reactive Oxygen Species/immunology , Uropathogenic Escherichia coli/immunology , Caspase 1/immunology , Humans , Inflammasomes/immunology , Neutrophils/microbiology , Neutrophils/pathology
12.
Front Immunol ; 11: 583276, 2020.
Article in English | MEDLINE | ID: mdl-33363533

ABSTRACT

Testicular macrophages (TM) play a central role in maintaining testicular immune privilege and protecting spermatogenesis. Recent studies showed that their immunosuppressive properties are maintained by corticosterone in the testicular interstitial fluid, but the underlying molecular mechanisms are unknown. In this study, we treated mouse bone marrow-derived macrophages (BMDM) with corticosterone (50 ng/ml) and uncovered AMP-activated protein kinase (AMPK) activation as a critical event in M2 polarization at the phenotypic, metabolic, and cytokine production level. Primary TM exhibited remarkably similar metabolic and phenotypic features to corticosterone-treated BMDM, which were partially reversed by AMPK-inhibition. In a murine model of uropathogenic E. coli-elicited orchitis, intraperitoneal injection with corticosterone (0.1mg/day) increased the percentage of M2 TM in vivo, in a partially AMPK-dependent manner. This study integrates the influence of corticosterone on M2 macrophage metabolic pathways, phenotype, and function, and highlights a promising new avenue for the development of innovative therapeutics for orchitis patients.


Subject(s)
Corticosterone/immunology , Escherichia coli Infections/immunology , Immune Tolerance/immunology , Macrophages/immunology , Orchitis/immunology , AMP-Activated Protein Kinases/immunology , AMP-Activated Protein Kinases/metabolism , Animals , Corticosterone/pharmacology , Male , Mice , Mice, Inbred C57BL , Orchitis/metabolism , Phenotype , Testis , Uropathogenic Escherichia coli/immunology
13.
Sci Rep ; 10(1): 16258, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33004862

ABSTRACT

Uropathogenic Escherichia coli (UPEC) are common pathogens in urinary tract infections (UTIs), which show resistance to antibiotics. Therefore, there is a need for a vaccine to reduce susceptibility to the infection. In the present study, bioinformatics approaches were employed to predict the best B and T-cell epitopes of UPEC virulence proteins to develop a multiepitope vaccine candidate against UPEC. Then, the efficacy of the candidate was studied with and without Freund adjuvant. Using bioinformatics methods, 3 epitope-rich domains of IutA and FimH antigens were selected to construct the fusion. Molecular docking and Molecular dynamics (MD) simulation were employed to investigate in silico interaction between designed vaccine and Toll-like receptor 4 (TLR4). Our results showed that the levels of IgG and IgA antibodies were improved in the serum and mucosal samples of the vaccinated mice, and the IgG responses were maintained for at least 6 months. The fusion protein was also able to enhance the level of cytokines IFN.γ (Th1), IL.4 (Th2), and IL.17. In challenge experiments, all vaccine combinations showed high potency in the protection of the urinary tract even after 6 months post first injection. The present study indicates that the designed candidate is able to evoke strong protective responses which warrant further studies.


Subject(s)
Escherichia coli Infections/prevention & control , Escherichia coli Vaccines/therapeutic use , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/immunology , Animals , Computer Simulation , Cytokines/metabolism , Epitopes/immunology , Escherichia coli Infections/immunology , Escherichia coli Vaccines/immunology , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Toll-Like Receptor 4/metabolism , Urinary Tract Infections/immunology
14.
Front Immunol ; 11: 2115, 2020.
Article in English | MEDLINE | ID: mdl-33117332

ABSTRACT

The epididymis is an important male accessory sex organ where sperm motility and fertilization ability develop. When spermatozoa carrying foreign antigens enter the epididymis, the epididymis shows "immune privilege" to tolerate them. It is well-known that a tolerogenic environment exists in the caput epididymis, while pro-inflammatory circumstances prefer the cauda epididymis. This meticulously regulated immune environment not only protects spermatozoa from autoimmunity but also defends spermatozoa against pathogenic damage. Epididymitis is one of the common causes of male infertility. Up to 40% of patients suffer from permanent oligospermia or azoospermia. This is related to the immune characteristics of the epididymis itself. Moreover, epididymitis induced by different pathogenic microbial infections has different characteristics. This article elaborates on the distribution and immune response characteristics of epididymis immune cells, the role of epididymis epithelial cells (EECs), and the epididymis defense against different pathogenic infections (such as uropathogenic Escherichia coli, Chlamydia trachomatis, and viruses to provide therapeutic approaches for epididymitis and its subsequent fertility problems.


Subject(s)
Epididymis/immunology , Epididymitis/immunology , Spermatozoa/immunology , Activins/physiology , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Blood-Testis Barrier , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Defensins/physiology , Epididymitis/complications , Epididymitis/epidemiology , Epididymitis/microbiology , Escherichia coli Infections/immunology , Humans , Immune System/cytology , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Infertility, Male/etiology , Infertility, Male/immunology , Infertility, Male/microbiology , Male , Mice , Middle Aged , TGF-beta Superfamily Proteins/physiology , Uropathogenic Escherichia coli/immunology , Virus Diseases/immunology , Young Adult
15.
J Immunol ; 205(10): 2763-2777, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33055280

ABSTRACT

Bacterial prostatitis affects 1% of men, with increased incidence in the elderly. Acute bacterial prostatitis frequently progresses to chronicity, marked by recurrent episodes interspersed with asymptomatic periods of variable duration. Antibiotic treatment is standard of care; however, dissemination of antimicrobially resistant uropathogens threatens therapy efficacy. Thus, development of nonantibiotic-based approaches to treat chronic disease is a priority. Currently, why chronic prostatitis arises is unclear, as the immune response to prostate infection is incompletely understood. As 80% of prostatitis cases are caused by Gram-negative uropathogenic Escherichia coli (UPEC) or Gram-positive Enterococcus faecalis, we used a mouse transurethral instillation model to address the hypothesis that an innate immune response fails to develop following prostate infection with these uropathogens, leading to chronic disease. Surprisingly, infection induced robust proinflammatory cytokine expression and myeloid cell infiltration. Following a second infection, cytokine responses and innate cell infiltration were largely comparable to primary infection. Characteristic of memory responses, more lymphoid cells infiltrated the prostate in a second infection compared with a first, suggesting that adaptive immunity develops to eliminate the pathogens. Unexpectedly, bacterial burden in prostates challenged with either UPEC or E. faecalis was equal or greater than primary infection despite that a protective adaptive response to UPEC infection was evident in the bladder of the same animals. Our findings support that chronic or recurrent prostatitis develops despite strong innate immune responses and may be the result of a failure to develop immune memory to infection, pointing to actionable targets for immunotherapy.


Subject(s)
Escherichia coli Infections/immunology , Gram-Positive Bacterial Infections/immunology , Prostatitis/immunology , Urinary Tract Infections/immunology , Animals , Chronic Disease , Cytokines/metabolism , Disease Models, Animal , Enterococcus faecalis/immunology , Enterococcus faecalis/pathogenicity , Escherichia coli Infections/microbiology , Escherichia coli Infections/therapy , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/therapy , Humans , Immunity, Innate , Immunologic Memory , Immunotherapy , Male , Mice , Prostate/immunology , Prostate/microbiology , Prostatitis/microbiology , Prostatitis/therapy , Recurrence , Urinary Bladder/immunology , Urinary Bladder/microbiology , Urinary Tract Infections/microbiology , Urinary Tract Infections/therapy , Uropathogenic Escherichia coli/immunology , Uropathogenic Escherichia coli/pathogenicity
16.
Sci Rep ; 10(1): 12653, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32724079

ABSTRACT

Hemolysin expressing UPEC strains have been associated with severe advanced kidney pathologies, such as cystitis and pyelonephritis, which are associated with an inflammatory response. Macrophages play an important role in regulating an inflammatory response during a urinary tract infection. We have studied the role of purified recombinant α-hemolysin in inducing inflammatory responses and cell death in macrophages. Acylation at lysine residues through HlyC is known to activate proHlyA into a fully functional pore-forming toxin, HlyA. It was observed that active α-hemolysin (HlyA) induced cleavage of caspase-1 leading to the maturation of IL-1ß, while inactive α-hemolysin (proHlyA) failed to do so in THP-1 derived macrophages. HlyA also promotes deubiquitination, oligomerization, and activation of the NLRP3 inflammasome, which was found to be dependent on potassium efflux. We have also observed the co-localization of NLRP3 within mitochondria during HlyA stimulations. Moreover, blocking of potassium efflux improved the mitochondrial health in addition to a decreased inflammatory response. Our study demonstrates that HlyA stimulation caused perturbance in potassium homeostasis, which led to the mitochondrial dysfunction followed by an acute inflammatory response, resulting in cell death. However, the repletion of intracellular potassium stores could avoid HlyA induced macrophage cell death. The findings of this study will help to understand the mechanism of α-hemolysin induced inflammatory response and cell death.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli Proteins/pharmacology , Hemolysin Proteins/pharmacology , Inflammasomes/physiology , Mitochondria/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Uropathogenic Escherichia coli/immunology , Acylation , Cell Death , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Humans , Inflammasomes/drug effects , Inflammation/drug therapy , Inflammation/immunology , Inflammation/microbiology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Mitochondria/drug effects , Mitochondria/immunology , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Potassium/metabolism , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/metabolism
17.
mBio ; 11(2)2020 04 28.
Article in English | MEDLINE | ID: mdl-32345645

ABSTRACT

Urinary tract infections (UTI) affect half of all women at least once during their lifetime. The rise in the numbers of extended-spectrum beta-lactamase-producing strains and the potential for carbapenem resistance within uropathogenic Escherichia coli (UPEC), the most common causative agent of UTI, create an urgent need for vaccine development. Intranasal immunization of mice with UPEC outer membrane iron receptors FyuA, Hma, IreA, and IutA, conjugated to cholera toxin, provides protection in the bladder or kidneys under conditions of challenge with UPEC strain CFT073 or strain 536. On the basis of these data, we sought to optimize the vaccination route (intramuscular, intranasal, or subcutaneous) in combination with adjuvants suitable for human use, including aluminum hydroxide gel (alum), monophosphoryl lipid A (MPLA), unmethylated CpG synthetic oligodeoxynucleotides (CpG), polyinosinic:polycytidylic acid (polyIC), and mutated heat-labile E. coli enterotoxin (dmLT). Mice intranasally vaccinated with dmLT-IutA and dmLT-Hma displayed significant reductions in bladder colonization (86-fold and 32-fold, respectively), with 40% to 42% of mice having no detectable CFU. Intranasal vaccination of mice with CpG-IutA and polyIC-IutA significantly reduced kidney colonization (131-fold) and urine CFU (22-fold), respectively. dmLT generated the most consistently robust antibody response in intranasally immunized mice, while MPLA and alum produced greater concentrations of antigen-specific serum IgG with intramuscular immunization. On the basis of these results, we conclude that intranasal administration of Hma or IutA formulated with dmLT adjuvant provides the greatest protection from UPEC UTI. This report advances our progress toward a vaccine against uncomplicated UTI, which will significantly improve the quality of life for women burdened by recurrent UTI and enable better antibiotic stewardship.IMPORTANCE Urinary tract infections (UTI) are among the most common bacterial infection in humans, affecting half of all women at least once during their lifetimes. The rise in antibiotic resistance and health care costs emphasizes the need to develop a vaccine against the most common UTI pathogen, Escherichia coli Vaccinating mice intranasally with a detoxified heat-labile enterotoxin and two surface-exposed receptors, Hma or IutA, significantly reduced bacterial burden in the bladder. This work highlights progress in the development of a UTI vaccine formulated with adjuvants suitable for human use and antigens that encode outer membrane iron receptors required for infection in the iron-limited urinary tract.


Subject(s)
Administration, Intranasal , Escherichia coli Proteins/immunology , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/immunology , Vaccines/therapeutic use , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/immunology , Drug Administration Routes , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Escherichia coli Infections/therapy , Female , Humans , Immunization/methods , Mice , Receptors, Cell Surface/immunology , Urinary Tract Infections/microbiology , Urinary Tract Infections/therapy , Uropathogenic Escherichia coli/pathogenicity , Vaccination/methods , Vaccines/administration & dosage
18.
Hum Antibodies ; 28(2): 169-178, 2020.
Article in English | MEDLINE | ID: mdl-32116244

ABSTRACT

Uropathogenic E. coli is one of the major agents of urinary tract infection. Today, no effective treatment or vaccine against this infection is exist. Accordingly, in the present study, a genetic constrruct for inducing of cellular immune system was designed. At first, fimH gene from E. coli 35218 was amplified using PCR. PCR product inserted into pET23a expression vector and the recombinant vector was analysed by sequencing. The vector was transformed to E. coli strain Origami and the protein was expressed under the 1 mM IPTG. FimH was purified with Ni-NTA column and the purified protein was used for immunization of BALB/c. Two weeks after the last injection, lymphocyte proliferation assay was carried out. In addition, IL-4 and IFN-γ cytokines, total antibody serum, IgG1 and IgG2a isotypes were quantified. Finally, protection ability of the vaccine in bladder and kidney infection of mice was evaluated.The results indicated that cellular immune response has a main protective role against UTI and FimH, as a vaccine candidate, significantly increase lymphocyte proliferation, IFN-γ response and total antibody amount. Immunization of mice with FimH conferred effective protection of kidney and bladder against urinary tract infection by uropathogenic E. coli (P< 0.002). It can be concluded that, the current FimH will be valuable for more trying to prepare a new vaccine against UTI.


Subject(s)
Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/immunology , Fimbriae Proteins/genetics , Fimbriae Proteins/immunology , Immunity/immunology , Lectins/genetics , Lectins/immunology , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/immunology , Animals , Antibodies, Bacterial/immunology , Escherichia coli Infections/immunology , Female , Immunization/methods , Kidney/immunology , Kidney/microbiology , Lymphocytes/immunology , Lymphocytes/microbiology , Mice , Mice, Inbred BALB C , Urinary Bladder/immunology , Urinary Bladder/microbiology , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology
19.
J Ethnopharmacol ; 260: 112578, 2020 Oct 05.
Article in English | MEDLINE | ID: mdl-31962152

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The seeds of Vaccaria segetalis (Neck.) Garcke is used for the treatment of urinary diseases in Traditional Chinese Medicine according to the Chinese Pharmacopoeia. Crude polysaccharides and the aqueous extract from the seeds of V. segetalis (SVCP) were proved to be effective on treating benign prostatic hyperplasia. AIM OF THE STUDY: The aim of this study was to test the effects of SVCP on urinary tract infection (UTI) induced by uropathogenic Escherichia coli (UPEC) strain CFT073 in the rat model and to investigate the underlying mechanisms. MATERIALS AND METHODS: A rat UTI model was established with the infection of UPEC strain CFT073. After oral administration of SVCP, the urinalysis and histological examination were evaluated. The levels of pro-inflammatory cytokines, procalcitonin (PCT) and polymeric Ig receptor (PIGR) were used to test the effects of SVCP on host immunity. The mRNA level of PapG in CFT073 was used to test the influence of SVCP on virulence factor. The effects of SVCP on the inhibition of bacterial adhesion were evaluated with mice UTI model. RESULTS: In the rat UTI model, the levels of bacterial load, white blood cells (WBC) and red blood cells (RBC) in urine and the pathological injury in the bladder were significantly up-regulated, the expression of PIGR in kidney was down-regulated, no significant change was observed on the pro-inflammatory cytokines in urine. After oral administration of SVCP for 3 days, the levels of bacterial load, WBC and RBC in urine were significantly decreased, the pathological injury in the bladder were remarkably inhibited. The expression of IL-6, IL-8 in urine and PIGR in kidney were significantly up-regulated by SVCP (200 mg/kg). SVCP showed no effect on the concentration of PCT in serum. SVCP failed to down-regulate the mRNA level of PapG in CFT073. In the mice UTI model, pre-treatment of SVCP failed to inhibit the intracellular bacterial load in the bladder. CONCLUSIONS: The therapeutic effects of SVCP on treating UTIs might result from the up-regulation of innate immunity in the kidney. SVCP can be used as an alternative therapeutic agent for UTIs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli Infections/prevention & control , Immunity, Innate/drug effects , Immunologic Factors/pharmacology , Kidney/drug effects , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Seeds , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/drug effects , Vaccaria , Animals , Anti-Bacterial Agents/isolation & purification , Bacterial Adhesion/drug effects , Bacterial Load , Cytokines/metabolism , Disease Models, Animal , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Female , Host-Pathogen Interactions , Immunologic Factors/isolation & purification , Inflammation Mediators/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/microbiology , Mice, Inbred C3H , Mice, Inbred C57BL , Plant Extracts/isolation & purification , Polysaccharides/isolation & purification , Rats, Sprague-Dawley , Seeds/chemistry , Signal Transduction , Urinary Bladder/drug effects , Urinary Bladder/microbiology , Urinary Tract Infections/immunology , Urinary Tract Infections/metabolism , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/immunology , Uropathogenic Escherichia coli/pathogenicity , Vaccaria/chemistry , Virulence/drug effects
20.
Am J Physiol Renal Physiol ; 318(2): F468-F474, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31841391

ABSTRACT

Acute pyelonephritis is frequently associated with metabolic acidosis. We previously reported that metabolic acidosis stimulates expression of hypoxia-inducible factor (HIF)-1α-induced target genes such as stromal derived factor-1 and cathelicidin, an antimicrobial peptide. Since the collecting duct (CD) plays a pivotal role in regulating acid-base homeostasis and is the first nephron segment encountered by an ascending microbial infection, we examined the contribution of HIF-1α to innate immune responses elicited by acid loading of an M-1 immortalized mouse CD cell line. Acid loading of confluent M-1 cells was achieved by culture in pH 6.8 medium supplemented with 5-(N-ethyl-N-isopropyl)-amiloride to block Na+/H+ exchange activity for 24 h. Acid loading induced antimicrobial peptide [cathelicidin and ß-defensin (Defb2 and Defb26)] mRNA expression and M-1 cell resistance to uropathogenic Escherichia coli infection to an extent similar to that obtained by inhibition of HIF prolyl hydroxylases, which promote HIF-1α protein degradation. The effect of acid loading on M-1 cell resistance to uropathogenic E. coli infection was reduced by inhibition of HIF-1α (PX-478), and, in combination with prolyl hydroxylase inhibitors, acidosis did not confer additional resistance. Thus, metabolic stress of acidosis triggers HIF-1α-dependent innate immune responses in CD (M-1) cells. Whether pharmacological stabilization of HIF prevents or ameliorates pyelonephritis in vivo warrants further investigation.


Subject(s)
Acidosis/metabolism , Antimicrobial Cationic Peptides/metabolism , Escherichia coli Infections/prevention & control , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Tubules, Collecting/metabolism , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/pathogenicity , Acidosis/immunology , Animals , Antimicrobial Cationic Peptides/genetics , Cell Line , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Immunity, Innate , Kidney Tubules, Collecting/immunology , Kidney Tubules, Collecting/microbiology , Mice , Prolyl Hydroxylases/metabolism , Protein Stability , Signal Transduction , Up-Regulation , Urinary Tract Infections/immunology , Urinary Tract Infections/metabolism , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/immunology , beta-Defensins/metabolism , Cathelicidins
SELECTION OF CITATIONS
SEARCH DETAIL
...