Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
PLoS One ; 17(8): e0269140, 2022.
Article in English | MEDLINE | ID: mdl-35980963

ABSTRACT

Chronic pelvic pain conditions such as interstitial cystitis/bladder pain syndrome (IC/BPS) remain clinical and mechanistic enigmas. Microglia are resident immune cells of the central nervous system (CNS) that respond to changes in the gut microbiome, and studies have linked microglial activation to acute and chronic pain in a variety of models, including pelvic pain. We have previously reported that mice deficient for the lipase acyloxyacyl hydrolase (AOAH) develop pelvic allodynia and exhibit symptoms, comorbidities, and gut dysbiosis mimicking IC/BPS. Here, we assessed the role of AOAH in microglial activation and pelvic pain. RNAseq analyses using the ARCHS4 database and confocal microscopy revealed that AOAH is highly expressed in wild type microglia but at low levels in astrocytes, suggesting a functional role for AOAH in microglia. Pharmacologic ablation of CNS microglia with PLX5622 resulted in decreased pelvic allodynia in AOAH-deficient mice and resurgence of pelvic pain upon drug washout. Skeletal analyses revealed that AOAH-deficient mice have an activated microglia morphology in the medial prefrontal cortex and paraventricular nucleus, brain regions associated with pain modulation. Because microglia express Toll-like receptors and respond to microbial components, we also examine the potential role of dysbiosis in microglial activation. Consistent with our hypothesis of microglia activation by leakage of gut microbes, we observed increased serum endotoxins in AOAH-deficient mice and increased activation of cultured BV2 microglial cells by stool of AOAH-deficient mice. Together, these findings demonstrate a role for AOAH in microglial modulation of pelvic pain and thus identify a novel therapeutic target for IC/BPS.


Subject(s)
Cystitis, Interstitial , Animals , Carboxylic Ester Hydrolases , Dysbiosis , Hyperalgesia , Mice , Mice, Inbred C57BL , Microglia , Pelvic Pain
2.
Am J Physiol Regul Integr Comp Physiol ; 321(3): R396-R412, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34318715

ABSTRACT

Dysbiosis of gut microbiota is associated with many pathologies, yet host factors modulating microbiota remain unclear. Interstitial cystitis/bladder pain syndrome (IC/BPS) is a debilitating condition of chronic pelvic pain often with comorbid urinary dysfunction and anxiety/depression, and recent studies find fecal dysbiosis in patients with IC/BPS. We identified the locus encoding acyloxyacyl hydrolase, Aoah, as a modulator of pelvic pain severity in a murine IC/BPS model. AOAH-deficient mice spontaneously develop rodent correlates of pelvic pain, increased responses to induced pelvic pain models, voiding dysfunction, and anxious/depressive behaviors. Here, we report that AOAH-deficient mice exhibit dysbiosis of gastrointestinal (GI) microbiota. AOAH-deficient mice exhibit an enlarged cecum, a phenotype long associated with germ-free rodents, and a "leaky gut" phenotype. AOAH-deficient ceca showed altered gene expression consistent with inflammation, Wnt signaling, and urologic disease. 16S sequencing of stool revealed altered microbiota in AOAH-deficient mice, and GC-MS identified altered metabolomes. Cohousing AOAH-deficient mice with wild-type mice resulted in converged microbiota and altered predicted metagenomes. Cohousing also abrogated the pelvic pain phenotype of AOAH-deficient mice, which was corroborated by oral gavage of AOAH-deficient mice with stool slurry of wild-type mice. Converged microbiota also alleviated comorbid anxiety-like behavior in AOAH-deficient mice. Oral gavage of AOAH-deficient mice with anaerobes cultured from IC/BPS stool resulted in exacerbation of pelvic allodynia. Together, these data indicate that AOAH is a host determinant of normal gut microbiota, and dysbiosis associated with AOAH deficiency contributes to pelvic pain. These findings suggest that the gut microbiome is a potential therapeutic target for IC/BPS.


Subject(s)
Carboxylic Ester Hydrolases , Cystitis, Interstitial , Gastrointestinal Microbiome , Pelvic Pain , Animals , Humans , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Cystitis, Interstitial/metabolism , Disease Models, Animal , Dysbiosis/complications , Dysbiosis/metabolism , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Inflammation/metabolism , Pelvic Pain/metabolism , Pelvic Pain/physiopathology , Urinary Bladder/metabolism , Mice
3.
PLoS One ; 15(9): e0235384, 2020.
Article in English | MEDLINE | ID: mdl-32925915

ABSTRACT

Interstitial cystitis/bladder pain syndrome (IC) is a debilitating condition of chronic pelvic pain with unknown etiology. Recently, we used a genetic approach in a murine model of IC to identify the lipase acyloxyacyl hydrolase (AOAH) as a modulator of pelvic pain. We found that AOAH-deficient mice have elevated pelvic pain responses, and AOAH immunoreactivity was detected along the bladder-brain axis. Lipidomic analyses identified arachidonic acid (AA) and its metabolite PGE2 as significantly elevated in the sacral spinal cord of AOAH-deficient mice, suggesting AA is a substrate for AOAH. Here, we quantified the effects of AOAH on phospholipids containing AA. Spinal cord lipidomics revealed increased AA-containing phosphatidylcholine in AOAH-deficient mice and concomitantly decreased AA-phosphatidylethanolamine, consistent with decreased CoA-independent transferase activity (CoIT). Overexpression of AOAH in cell cultures similarly altered distribution of AA in phospholipid pools, promoted AA incorporation, and resulted in decreased membrane fluidity. Finally, administration of a PGE2 receptor antagonist reduced pelvic pain in AOAH-deficient mice. Together, these findings suggest that AOAH represents a potential CoA-independent AA transferase that modulates CNS pain pathways at the level of phospholipid metabolism.


Subject(s)
Arachidonic Acid/metabolism , Carboxylic Ester Hydrolases/metabolism , Cystitis, Interstitial/metabolism , Pelvic Pain/metabolism , Phospholipids/metabolism , Animals , Cystitis, Interstitial/complications , Disease Models, Animal , Female , Mice, Inbred C57BL , Pelvic Pain/complications , Urinary Bladder/metabolism
4.
Am J Physiol Renal Physiol ; 318(4): F1006-F1016, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32003596

ABSTRACT

Corticotropin-releasing factor (CRF) regulates diverse physiological functions, including bladder control. We recently reported that Crf expression is under genetic control of Aoah, the locus encoding acyloxyacyl hydrolase (AOAH), suggesting that AOAH may also modulate voiding. Here, we examined the role of AOAH in bladder function. AOAH-deficient mice exhibited enlarged bladders relative to wild-type mice and had decreased voiding frequency and increased void volumes. AOAH-deficient mice had increased nonvoiding contractions and increased peak voiding pressure in awake cystometry. AOAH-deficient mice also exhibited increased bladder permeability and higher neuronal firing rates of bladder afferents in response to stretch. In wild-type mice, AOAH was expressed in bladder projecting neurons and colocalized in CRF-expressing neurons in Barrington's nucleus, an important brain area for voiding behavior, and Crf was elevated in Barrington's nucleus of AOAH-deficient mice. We had previously identified aryl hydrocarbon receptor (AhR) and peroxisome proliferator-activated receptor-γ as transcriptional regulators of Crf, and conditional knockout of AhR or peroxisome proliferator-activated receptor-γ in Crf-expressing cells restored normal voiding in AOAH-deficient mice. Finally, an AhR antagonist improved voiding in AOAH-deficient mice. Together, these data demonstrate that AOAH regulates bladder function and that the AOAH-Crf axis is a therapeutic target for treating voiding dysfunction.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Neurons/enzymology , Urinary Bladder/innervation , Urination Disorders/enzymology , Urination , Urodynamics , Animals , Azo Compounds/pharmacology , Barrington's Nucleus/metabolism , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carboxylic Ester Hydrolases/deficiency , Carboxylic Ester Hydrolases/genetics , Corticotropin-Releasing Hormone/metabolism , Female , Male , Mice, Inbred C57BL , Muscle Contraction , Neurons/drug effects , PPAR gamma/genetics , PPAR gamma/metabolism , Pressure , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Urinary Bladder/drug effects , Urination/drug effects , Urination Disorders/drug therapy , Urination Disorders/genetics , Urination Disorders/physiopathology , Urodynamics/drug effects
5.
Am J Physiol Regul Integr Comp Physiol ; 317(2): R289-R300, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31017816

ABSTRACT

Corticotropin-releasing factor (CRF) regulates stress responses, and aberrant CRF signals are associated with depressive disorders. Crf expression is responsive to arachidonic acid (AA), where CRF is released from the hypothalamic paraventricular nucleus (PVN) to initiate the hypothalamic-pituitary-adrenal axis, culminating in glucocorticoid stress hormone release. Despite this biological and clinical significance, Crf regulation is unclear. Here, we report that acyloxyacyl hydrolase, encoded by Aoah, is expressed in the PVN, and Aoah regulates Crf through the aryl hydrocarbon receptor (AhR). We previously showed that AOAH-deficient mice mimicked interstitial cystitis/bladder pain syndrome, a condition frequently associated with comorbid anxiety and depression. With the use of novelty-suppressed feeding and sucrose preference assays to quantify rodent correlates of anxiety/depression, AOAH-deficient mice exhibited depressive behaviors. AOAH-deficient mice also had increased CNS AA, increased Crf expression in the PVN, and elevated serum corticosterone, consistent with dysfunction of the hypothalamic-pituitary-adrenal axis. The human Crf promoter has putative binding sites for AhR and peroxisome proliferator-activated receptor (PPARγ). PPARγ did not affect AA-dependent Crf expression in vitro, and conditional Pparγ knockout did not alter the AOAH-deficient depressive phenotype, despite previous studies implicating PPARγ as a therapeutic target for depression. In contrast, Crf induction was mediated by AhR binding sites in vitro and increased by AhR overexpression. Furthermore, conditional Ahr knockout rescued the depressive phenotype of AOAH-deficient mice. Finally, an AhR antagonist rescued the AOAH-deficient depressive phenotype. Together, our results demonstrate that Aoah is a novel genetic regulator of Crf mediated through AhR, and AhR is a therapeutic target for depression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Carboxylic Ester Hydrolases/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Stress, Psychological/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Corticosterone/blood , Corticotropin-Releasing Hormone/metabolism , Female , Gene Expression Regulation/physiology , Humans , Hypothalamo-Hypophyseal System/metabolism , Mice, Transgenic , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/metabolism , Receptors, Aryl Hydrocarbon/metabolism
6.
Sci Rep ; 8(1): 7188, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29739958

ABSTRACT

The etiology of chronic pelvic pain syndromes remains unknown. In a murine urinary tract infection (UTI) model, lipopolysaccharide of uropathogenic E. coli and its receptor TLR4 are required for post-UTI chronic pain development. However, downstream mechanisms of post-UTI chronic pelvic pain remain unclear. Because the TRPV1 and MCP-1/CCR2 pathways are implicated in chronic neuropathic pain, we explored their role in post-UTI chronic pain. Mice were infected with the E. coli strain SΦ874, known to produce chronic allodynia, and treated with the TRPV1 antagonist capsazepine. Mice treated with capsazepine at the time of SΦ874 infection failed to develop chronic allodynia, whereas capsazepine treatment of mice at two weeks following SΦ874 infection did not reduce chronic allodynia. TRPV1-deficient mice did not develop chronic allodynia either. Similar results were found using novelty-suppressed feeding (NSF) to assess depressive behavior associated with neuropathic pain. Imaging of reporter mice also revealed induction of MCP-1 and CCR2 expression in sacral dorsal root ganglia following SΦ874 infection. Treatment with a CCR2 receptor antagonist at two weeks post-infection reduced chronic allodynia. Taken together, these results suggest that TRPV1 has a role in the establishment of post-UTI chronic pain, and CCR2 has a role in maintenance of post-UTI chronic pain.


Subject(s)
Chemokine CCL2/genetics , Chronic Pain/metabolism , Hyperalgesia/metabolism , Pelvic Pain/metabolism , Receptors, CCR2/genetics , TRPV Cation Channels/genetics , Urinary Tract Infections/metabolism , Animals , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Chemokine CCL2/metabolism , Chronic Pain/genetics , Chronic Pain/microbiology , Chronic Pain/physiopathology , Disease Models, Animal , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/microbiology , Ganglia, Spinal/physiopathology , Gene Expression Regulation , Hyperalgesia/genetics , Hyperalgesia/microbiology , Hyperalgesia/physiopathology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Pelvic Pain/genetics , Pelvic Pain/microbiology , Pelvic Pain/physiopathology , Receptors, CCR2/metabolism , Signal Transduction , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology , Urinary Tract Infections/physiopathology , Uropathogenic Escherichia coli/chemistry , Uropathogenic Escherichia coli/pathogenicity , Uropathogenic Escherichia coli/physiology
7.
Am J Physiol Regul Integr Comp Physiol ; 314(3): R353-R365, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29118019

ABSTRACT

Chronic pelvic pain causes significant patient morbidity and is a challenge to clinicians. Using a murine neurogenic cystitis model that recapitulates key aspects of interstitial cystitis/bladder pain syndrome (IC), we recently showed that pseudorabies virus (PRV) induces severe pelvic allodynia in BALB/c mice relative to C57BL/6 mice. Here, we report that a quantitative trait locus (QTL) analysis of PRV-induced allodynia in F2CxB progeny identified a polymorphism on chromosome 13, rs6314295 , significantly associated with allodynia (logarithm of odds = 3.11). The nearby gene encoding acyloxyacyl hydrolase ( Aoah) was induced in the sacral spinal cord of PRV-infected mice. AOAH-deficient mice exhibited increased vesicomotor reflex in response to bladder distension, consistent with spontaneous bladder hypersensitivity, and increased pelvic allodynia in neurogenic cystitis and postbacterial chronic pain models. AOAH deficiency resulted in greater bladder pathology and tumor necrosis factor production in PRV neurogenic cystitis, markers of increased bladder mast cell activation. AOAH immunoreactivity was detectable along the bladder-brain axis, including in brain sites previously correlated with human chronic pelvic pain. Finally, AOAH-deficient mice had significantly higher levels of bladder vascular endothelial growth factor, an emerging marker of chronic pelvic pain in humans. These findings indicate that AOAH modulates pelvic pain severity, suggesting that allelic variation in Aoah influences pelvic pain in IC.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Cystitis, Interstitial/enzymology , Escherichia coli Infections/enzymology , Hyperalgesia/enzymology , Pelvic Pain/enzymology , Pseudorabies/enzymology , Urinary Bladder/innervation , Urinary Tract Infections/enzymology , Animals , Behavior, Animal , Carboxylic Ester Hydrolases/deficiency , Carboxylic Ester Hydrolases/genetics , Cystitis, Interstitial/genetics , Cystitis, Interstitial/physiopathology , Cystitis, Interstitial/psychology , Disease Models, Animal , Escherichia coli Infections/genetics , Escherichia coli Infections/physiopathology , Escherichia coli Infections/psychology , Female , Genetic Predisposition to Disease , Hyperalgesia/genetics , Hyperalgesia/physiopathology , Hyperalgesia/psychology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pain Perception , Pain Threshold , Pelvic Pain/genetics , Pelvic Pain/physiopathology , Phenotype , Pseudorabies/genetics , Pseudorabies/physiopathology , Pseudorabies/psychology , Quantitative Trait Loci , Severity of Illness Index , Tumor Necrosis Factor-alpha/metabolism , Urinary Bladder/metabolism , Urinary Tract Infections/genetics , Urinary Tract Infections/physiopathology , Urinary Tract Infections/psychology , Vascular Endothelial Growth Factor A/metabolism
8.
Infect Immun ; 84(11): 3131-3140, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27528276

ABSTRACT

Uropathogenic Escherichia coli (UPEC) accounts for 80 to 90% of urinary tract infections (UTI), and the increasing rate of antibiotic resistance among UPEC isolates reinforces the need for vaccines to prevent UTIs and recurrent infections. Previous studies have shown that UPEC isolate NU14 suppresses proinflammatory NF-κB-dependent cytokines (D. J. Klumpp, A. C. Weiser, S. Sengupta, S. G. Forrestal, R. A. Batler, and A. J. Schaeffer, Infect Immun 69:6689-6695, 2001, http://dx.doi.org/10.1128/IAI.69.11.6689-6695.2001; B. K. Billips, A. J. Schaeffer, and D. J. Klumpp, Infect Immun 76:3891-3900, 2008, http://dx.doi.org/10.1128/IAI.00069-08). However, modification of lipopolysaccharide (LPS) structure by deleting the O-antigen ligase gene (waaL) enhanced proinflammatory cytokine secretion. Vaccination with the ΔwaaL mutant diminished NU14 reservoirs and protected against subsequent infections. Therefore, we hypothesized that LPS structural determinants shape immune responses. We evaluated the contribution of LPS domains to urovirulence corresponding to the inner core (waaP, waaY, and rfaQ), outer core (rfaG), and O-antigen (waaL, wzzE, and wzyE). Deletion of waaP, waaY, and rfaG attenuated adherence to urothelial cells in vitro In a murine UTI model, the ΔrfaG mutant had the most severe defect in colonization. The mutation of rfaG, waaL, wzzE, and wzyE resulted in an inability to form reservoirs in mouse bladders. Infection with the LPS mutant panel resulted in various levels of urinary myeloperoxidase. Since the ΔwaaL mutant promoted Th1-associated adaptive responses in previous studies (B. K. Billips, R. E. Yaggie, J. P. Cashy, A. J. Schaeffer, and D. J. Klumpp, J Infect Dis 200:263-272, 2009, http://dx.doi.org/10.1086/599839), we assessed NU14 for Th2-associated cytokines. We found NU14 infection stimulated TLR4-dependent bladder interleukin-33 (IL-33) production. Inoculation with rfaG, waaL, wzzE, and wzyE mutants showed decreased IL-33 production. We quantified antigen-specific antibodies after infection and found significantly increased IgE and IgG1 in ΔwaaP mutant-infected mice. Our studies show LPS structural constituents mediate multiple aspects of the UPEC life cycle, including the ability to acutely colonize bladders, form reservoirs, and evoke innate and adaptive immune responses.


Subject(s)
Escherichia coli Infections , Lipopolysaccharides/physiology , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/pathogenicity , Virulence/physiology , Adaptive Immunity/physiology , Animals , Disease Models, Animal , Escherichia coli Infections/immunology , Female , Immunity, Innate/physiology , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , O Antigens/immunology , Peroxidase/metabolism , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli/genetics
9.
Sci Rep ; 6: 26083, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27188581

ABSTRACT

Interstitial cystitis/bladder pain syndrome (IC) is associated with significant morbidity, yet underlying mechanisms and diagnostic biomarkers remain unknown. Pelvic organs exhibit neural crosstalk by convergence of visceral sensory pathways, and rodent studies demonstrate distinct bacterial pain phenotypes, suggesting that the microbiome modulates pelvic pain in IC. Stool samples were obtained from female IC patients and healthy controls, and symptom severity was determined by questionnaire. Operational taxonomic units (OTUs) were identified by16S rDNA sequence analysis. Machine learning by Extended Random Forest (ERF) identified OTUs associated with symptom scores. Quantitative PCR of stool DNA with species-specific primer pairs demonstrated significantly reduced levels of E. sinensis, C. aerofaciens, F. prausnitzii, O. splanchnicus, and L. longoviformis in microbiota of IC patients. These species, deficient in IC pelvic pain (DIPP), were further evaluated by Receiver-operator characteristic (ROC) analyses, and DIPP species emerged as potential IC biomarkers. Stool metabolomic studies identified glyceraldehyde as significantly elevated in IC. Metabolomic pathway analysis identified lipid pathways, consistent with predicted metagenome functionality. Together, these findings suggest that DIPP species and metabolites may serve as candidates for novel IC biomarkers in stool. Functional changes in the IC microbiome may also serve as therapeutic targets for treating chronic pelvic pain.


Subject(s)
Bacteria/classification , Biomarkers/analysis , Cystitis, Interstitial/pathology , Feces/chemistry , Feces/microbiology , Metabolome , Urinary Bladder/pathology , Adult , Bacteria/genetics , Cluster Analysis , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Female , Humans , Metagenomics , Middle Aged , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Surveys and Questionnaires , Young Adult
10.
PLoS One ; 9(11): e109321, 2014.
Article in English | MEDLINE | ID: mdl-25405579

ABSTRACT

Urinary tract infections (UTI) account for approximately 8 million clinic visits annually with symptoms that include acute pelvic pain, dysuria, and irritative voiding. Empiric UTI management with antimicrobials is complicated by increasing antimicrobial resistance among uropathogens, but live biotherapeutics products (LBPs), such as asymptomatic bacteriuria (ASB) strains of E. coli, offer the potential to circumvent antimicrobial resistance. Here we evaluated ASB E. coli as LBPs, relative to ciprofloxacin, for efficacy against infection and visceral pain in a murine UTI model. Visceral pain was quantified as tactile allodynia of the pelvic region in response to mechanical stimulation with von Frey filaments. Whereas ciprofloxacin promoted clearance of uropathogenic E. coli (UPEC), it did not reduce pelvic tactile allodynia, a measure of visceral pain. In contrast, ASB E. coli administered intravesically or intravaginally provided comparable reduction of allodynia similar to intravesical lidocaine. Moreover, ASB E. coli were similarly effective against UTI allodynia induced by Proteus mirabilis, Enterococccus faecalis and Klebsiella pneumoniae. Therefore, ASB E. coli have anti-infective activity comparable to the current standard of care yet also provide superior analgesia. These studies suggest that ASB E. coli represent novel LBPs for UTI symptoms.


Subject(s)
Asymptomatic Infections , Bacteriuria/therapy , Biological Therapy , Escherichia coli/isolation & purification , Animals , Bacteriuria/microbiology , Female , Mice , Mice, Inbred C57BL
11.
PLoS One ; 7(8): e41273, 2012.
Article in English | MEDLINE | ID: mdl-22899994

ABSTRACT

The molecular initiators of infection-associated pain are not understood. We recently found that uropathogenic E. coli (UPEC) elicited acute pelvic pain in murine urinary tract infection (UTI). UTI pain was due to E. coli lipopolysaccharide (LPS) and its receptor, TLR4, but pain was not correlated with inflammation. LPS is known to drive inflammation by interactions between the acylated lipid A component and TLR4, but the function of the O-antigen polysaccharide in host responses is unknown. Here, we examined the role of O-antigen in pain using cutaneous hypersensitivity (allodynia) to quantify pelvic pain behavior and using sacral spinal cord excitability to quantify central nervous system manifestations in murine UTI. A UPEC mutant defective for O-antigen biosynthesis induced chronic allodynia that persisted long after clearance of transient infections, but wild type UPEC evoked only acute pain. E. coli strains lacking O-antigen gene clusters had a chronic pain phenotype, and expressing cloned O-antigen gene clusters altered the pain phenotype in a predictable manner. Chronic allodynia was abrogated in TLR4-deficient mice, but inflammatory responses in wild type mice were similar among E. coli strains spanning a wide range of pain phenotypes, suggesting that O-antigen modulates pain independent of inflammation. Spinal cords of mice with chronic allodynia exhibited increased spontaneous firing and compromised short-term depression, consistent with centralized pain. Taken together, these findings suggest that O-antigen functions as a rheostat to modulate LPS-associated pain. These observations have implications for an infectious etiology of chronic pain and evolutionary modification of pathogens to alter host behaviors.


Subject(s)
Escherichia coli Infections/immunology , O Antigens/immunology , Pelvic Pain/etiology , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , Animals , Female , Hyperalgesia/immunology , Hyperalgesia/microbiology , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharides/immunology , Mice , Pelvic Pain/immunology , Phenotype , Spinal Cord/physiopathology , Toll-Like Receptor 4/metabolism
12.
J Infect Dis ; 201(8): 1240-9, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20225955

ABSTRACT

BACKGROUND: Pelvic pain is a major component of the morbidity associated with urinary tract infection (UTI), yet the molecular mechanisms underlying UTI-induced pain remain unknown. UTI pain mechanisms probably contrast with the clinical condition of asymptomatic bacteriuria (ASB), characterized by significant bacterial loads without lack symptoms. METHODS: A murine UTI model was used to compare pelvic pain behavior elicited by infection with uropathogenic Escherichia coli strain NU14 and ASB strain 83972. RESULTS: NU14-infected mice exhibited pelvic pain, whereas mice infected with 83972 did not exhibit pain, similar to patients infected with 83972. NU14-induced pain was not dependent on mast cells, not correlated with bacterial colonization or urinary neutrophils. UTI pain was not influenced by expression of type 1 pili, the bacterial adhesive appendages that induce urothelial apoptosis. However, purified NU14 lipopolysaccharide (LPS) induced Toll-like receptor 4 (TLR4)-dependent pain, whereas 83972 LPS induced no pain. Indeed, 83972 LPS attenuated the pain of NU14 infection, suggesting therapeutic potential. CONCLUSIONS: These data suggest a novel mechanism of infection-associated pain that is dependent on TLR4 yet independent of inflammation. Clinically, these findings also provide the rational for probiotic therapies that would minimize the symptoms of infection without reliance on empirical therapies that contribute to antimicrobial resistance.


Subject(s)
Host-Pathogen Interactions/physiology , Pelvic Pain/microbiology , Urinary Tract Infections/microbiology , Animals , Carrier State/microbiology , Carrier State/physiopathology , Escherichia coli Infections/microbiology , Escherichia coli Infections/physiopathology , Female , Lipopolysaccharides/physiology , Macrophages/physiology , Mast Cells/physiology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Pelvic Pain/physiopathology , Receptors, Immunologic/physiology , Toll-Like Receptor 4/physiology , Urinary Bladder Diseases/microbiology , Urinary Tract Infections/physiopathology , Uropathogenic Escherichia coli/physiology
13.
J Infect Dis ; 200(2): 263-72, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19522648

ABSTRACT

Uropathogenic Escherichia coli are the leading cause of urinary tract infection. We recently demonstrated that deletion of the O antigen ligase gene, waaL, from the uropathogenic E. coliisolate NU14 results in a strain that stimulates enhanced urothelial cytokine secretion. Because enhanced innate immune responses are of interest in vaccine development, we examined the therapeutic potential of NU14 DeltawaaL as a vaccine for urinary tract infection. NU14 DeltawaaL stimulated enhanced interleukin-6 secretion by mouse macrophages, compared with secretion by the wild type. Mice vaccinated via instillation into the bladder developed protective responses that prevented persistent colonization after bladder challenge with NU14, yet NU14 DeltawaaL failed to persistently colonize the mouse bladder. Inoculation with the vaccine strain protected mice against challenge with a broad range of clinical uropathogenic E. coli isolates and produced immunity that lasted 8 weeks. Therefore, NU14 DeltawaaL is a candidate live-attenuated vaccine for the treatment and prevention of acute and recurrent urinary tract infection by caused by uropathogenic E. coli.


Subject(s)
Bacterial Vaccines , Escherichia coli Infections/prevention & control , Escherichia coli/immunology , Urinary Tract Infections/prevention & control , Animals , Cell Line , Escherichia coli/genetics , Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Female , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Urinary Bladder/immunology , Urinary Bladder/microbiology , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , Vaccination , Vaccines, Attenuated
14.
PLoS Pathog ; 5(5): e1000415, 2009 May.
Article in English | MEDLINE | ID: mdl-19412341

ABSTRACT

Urinary tract infections are the second most common infectious disease in humans and are predominantly caused by uropathogenic E. coli (UPEC). A majority of UPEC isolates express the type 1 pilus adhesin, FimH, and cell culture and murine studies demonstrate that FimH is involved in invasion and apoptosis of urothelial cells. FimH initiates bladder pathology by binding to the uroplakin receptor complex, but the subsequent events mediating pathogenesis have not been fully characterized. We report a hitherto undiscovered signaling role for the UPIIIa protein, the only major uroplakin with a potential cytoplasmic signaling domain, in bacterial invasion and apoptosis. In response to FimH adhesin binding, the UPIIIa cytoplasmic tail undergoes phosphorylation on a specific threonine residue by casein kinase II, followed by an elevation of intracellular calcium. Pharmacological inhibition of these signaling events abrogates bacterial invasion and urothelial apoptosis in vitro and in vivo. Our studies suggest that bacteria-induced UPIIIa signaling is a critical mediator of bladder responses to insult by uropathogenic E. coli.


Subject(s)
Escherichia coli Infections/microbiology , Escherichia coli/pathogenicity , Membrane Glycoproteins/metabolism , Signal Transduction , Urinary Tract Infections/microbiology , Adhesins, Escherichia coli/metabolism , Animals , Apoptosis , Bacterial Adhesion , Calcium/metabolism , Casein Kinase II/metabolism , Cell Line, Transformed , Escherichia coli/physiology , Escherichia coli Proteins/metabolism , Female , Fimbriae Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Phosphorylation , Uroplakin III , Urothelium/cytology , Urothelium/metabolism
15.
J Biol Chem ; 279(48): 49948-55, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15385550

ABSTRACT

An 18-bp enhancer controls cell-specific expression of the calcitonin/calcitonin gene-related peptide gene. The enhancer is bound by a heterodimer of the bHLH-Zip protein USF-1 and -2 and a cell-specific factor from thyroid C cell lines. In this report we have identified the cell-specific factor as the forkhead protein Foxa2 (previously HNF-3beta). Binding of Foxa2 to the 18-bp enhancer was demonstrated using electrophoretic mobility shift assays. The cell-specific DNA-protein complex was selectively competed by a series of Foxa2 DNA binding sites, and the addition of Foxa2 antiserum supershifted the complex. Likewise, a complex similar to that seen with extracts from thyroid C cell lines was generated using an extract from heterologous cells expressing recombinant Foxa2. Interestingly, overexpression of Foxa2 activated the 18-bp enhancer in heterologous cells but only in the presence of the adjacent helix-loop-helix motif. Likewise, coexpression of USF proteins with Foxa2 yielded greater activation than by Foxa2 alone. Unexpectedly, Foxa2 overexpression repressed activity in the CA77 thyroid C cell line, suggesting that Foxa2 may interact with additional cofactors. The stimulatory role of Foxa2 at the calcitonin/calcitonin gene-related peptide gene enhancer was confirmed by short interfering RNA-mediated knockdown of Foxa2. As seen with Foxa2 overexpression, the effect of Foxa2 knockdown also required the adjacent helix-loop-helix motif. These results provide the first evidence for combinatorial control of gene expression by bHLH-Zip and forkhead proteins.


Subject(s)
Calcitonin Gene-Related Peptide/genetics , Calcitonin/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Calcitonin/metabolism , Calcitonin Gene-Related Peptide/metabolism , Gene Expression Regulation/physiology , Hepatocyte Nuclear Factor 3-beta , Humans , Rats , Upstream Stimulatory Factors
SELECTION OF CITATIONS
SEARCH DETAIL