Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
1.
FASEB J ; 34(9): 11641-11657, 2020 09.
Article in English | MEDLINE | ID: mdl-32654268

ABSTRACT

The tight junction (TJ) and barrier function of colonic epithelium is highly sensitive to ionizing radiation. We evaluated the effect of lysophosphatidic acid (LPA) and its analog, Radioprotein-1, on γ-radiation-induced colonic epithelial barrier dysfunction using Caco-2 and m-ICC12 cell monolayers in vitro and mice in vivo. Mice were subjected to either total body irradiation (TBI) or partial body irradiation (PBI-BM5). Intestinal barrier function was assessed by analyzing immunofluorescence localization of TJ proteins, mucosal inulin permeability, and plasma lipopolysaccharide (LPS) levels. Oxidative stress was analyzed by measuring protein thiol oxidation and antioxidant mRNA. In Caco-2 and m-ICC12 cell monolayers, LPA attenuated radiation-induced redistribution of TJ proteins, which was blocked by a Rho-kinase inhibitor. In mice, TBI and PBI-BM5 disrupted colonic epithelial tight junction and adherens junction, increased mucosal permeability, and elevated plasma LPS; TJ disruption by TBI was more severe in Lpar2-/- mice compared to wild-type mice. RP1, administered before or after irradiation, alleviated TBI and PBI-BM5-induced TJ disruption, barrier dysfunction, and endotoxemia accompanied by protein thiol oxidation and downregulation of antioxidant gene expression, cofilin activation, and remodeling of the actin cytoskeleton. These data demonstrate that LPAR2 receptor activation prevents and mitigates γ-irradiation-induced colonic mucosal barrier dysfunction and endotoxemia.


Subject(s)
Colon/radiation effects , Intestinal Mucosa/radiation effects , Radiation, Ionizing , Receptors, Lysophosphatidic Acid/genetics , Tight Junctions/radiation effects , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Adherens Junctions/radiation effects , Animals , Caco-2 Cells , Cell Line , Colon/drug effects , Colon/metabolism , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Intercellular Junctions/radiation effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lysophospholipids/pharmacology , Mice, Knockout , Permeability/drug effects , Permeability/radiation effects , Receptors, Lysophosphatidic Acid/metabolism , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism
2.
J Innate Immun ; 10(1): 14-29, 2018.
Article in English | MEDLINE | ID: mdl-29069656

ABSTRACT

Urinary tract infections (UTIs) mainly due to uropathogenic Escherichia coli (UPEC) are one of the most frequent complications in kidney-transplanted patients, causing significant morbidity. However, the mechanisms underlying UTI in renal grafts remain poorly understood. Here, we analysed the effects of the potent immunosuppressive agent cyclosporine A (CsA) on the activation of collecting duct cells that represent a preferential site of adhesion and translocation for UPEC. CsA induced the inhibition of lipopolysaccharide- induced activation of collecting duct cells due to the downregulation of the expression of TLR4 via the microRNA Let-7i. Using an experimental model of ascending UTI, we showed that the pretreatment of mice with CsA prior to infection induced a marked fall in cytokine production by collecting duct cells, neutrophil recruitment, and a dramatic rise of bacterial load, but not in infected TLR4-defective mice kidneys. This effect was also observed in CsA-treated infected kidneys, where the expression of Let-7i was increased. Treatment with a synthetic Let-7i mimic reproduced the effects of CsA. Conversely, pretreatment with an anti-Let-7i antagonised the effects of CsA and rescued the innate immune response of collecting duct cells against UPEC. Thus, the utilisation of an anti-Let-7i during kidney transplantation may protect CsA-treated patients from ascending bacterial infection.


Subject(s)
Cyclosporine/therapeutic use , Escherichia coli Infections/drug therapy , Immunosuppressive Agents/therapeutic use , Kidney Tubules, Collecting/drug effects , MicroRNAs/genetics , Urinary Tract Infections/drug therapy , Uropathogenic Escherichia coli/physiology , Animals , Cells, Cultured , Female , Humans , Immunity, Innate , Kidney Tubules, Collecting/microbiology , Kidney Tubules, Collecting/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
3.
Cell Microbiol ; 19(8)2017 08.
Article in English | MEDLINE | ID: mdl-28296078

ABSTRACT

Botulinum neurotoxins (BoNTs) are responsible for severe flaccid paralysis by inhibiting the release of acetylcholine at the neuromuscular junctions. BoNT type B (BoNT/B) most often induces mild forms of botulism with predominant dysautonomic symptoms. In food borne botulism and botulism by intestinal colonisation such as infant botulism, which are the most frequent naturally acquired forms of botulism, the digestive tract is the main entry route of BoNTs into the organism. We previously showed that BoNT/B translocates through mouse intestinal barrier by an endocytosis-dependent mechanism and subsequently targets neuronal cells, mainly cholinergic neurons, in the intestinal mucosa and musculosa. Here, we investigated the entry pathway of BoNT/B using fluorescent C-terminal domain of the heavy chain (HcB), which is involved in the binding to specific receptor(s) and entry process into target cells. While the combination of gangliosides GD1a /GD1b /GT1b and synaptotagmin I and to a greater extent synaptotagmin II constitutes the functional HcB receptor on NG108-15 neuronal cells, HcB only uses the gangliosides GD1a /GD1b /GT1b to efficiently bind to m-ICcl2 intestinal cells. HcB enters both cell types by a dynamin-dependent endocytosis, which is efficiently prevented by Dynasore, a dynamin inhibitor, and reaches a common early endosomal compartment labeled by early endosome antigen (EEA1). In contrast to neuronal cells, HcB uses a Cdc42-dependent pathway to enter intestinal cells. Then, HcB is transported to late endosomes in neuronal cells, whereas it exploits a nonacidified pathway from apical to basal lateral side of m-ICcl2 cells supporting a transcytotic route in epithelial intestinal cells.


Subject(s)
Botulinum Toxins, Type A/metabolism , Endocytosis , Epithelial Cells/metabolism , Neurons/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , Mice
4.
Nat Commun ; 7: 13636, 2016 11 28.
Article in English | MEDLINE | ID: mdl-27892464

ABSTRACT

Heritable nephrogenic diabetes insipidus (NDI) is characterized by defective urine concentration mechanisms in the kidney, which are mainly caused by loss-of-function mutations in the vasopressin type 2 receptor. For the treatment of heritable NDI, novel strategies that bypass the defective vasopressin type 2 receptor are required to activate the aquaporin-2 (AQP2) water channel. Here we show that Wnt5a regulates AQP2 protein expression, phosphorylation and trafficking, suggesting that Wnt5a is an endogenous ligand that can regulate AQP2 without the activation of the classic vasopressin/cAMP signalling pathway. Wnt5a successfully increases the apical membrane localization of AQP2 and urine osmolality in an NDI mouse model. We also demonstrate that calcineurin is a key regulator of Wnt5a-induced AQP2 activation without affecting intracellular cAMP level and PKA activity. The importance of calcineurin is further confirmed with its activator, arachidonic acid, which shows vasopressin-like effects underlining that calcineurin activators may be potential therapeutic targets for heritable NDI.


Subject(s)
Aquaporin 2/genetics , Calcineurin/metabolism , Gene Expression Regulation , Kidney/metabolism , Signal Transduction , Wnt-5a Protein/metabolism , Animals , Aquaporin 2/metabolism , Arachidonic Acid/pharmacology , Calcium/metabolism , Calcium Signaling/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Insipidus, Nephrogenic/metabolism , Diabetes Insipidus, Nephrogenic/pathology , Diabetes Insipidus, Nephrogenic/urine , Disease Models, Animal , Dishevelled Proteins/metabolism , Gene Expression Regulation/drug effects , Male , Mice, Inbred C57BL , Models, Biological , Osmolar Concentration , Permeability , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Water , beta Catenin/metabolism
5.
Sci Rep ; 6: 18710, 2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26732173

ABSTRACT

The With No lysine [K] (WNK)-Ste20-related proline/alanine-rich kinase (SPAK)/oxidative stress-responsive kinase 1 (OSR1) pathway has been reported to be a crucial signaling pathway for triggering pseudohypoaldosteronism type II (PHAII), an autosomal dominant hereditary disease that is characterized by hypertension. However, the molecular mechanism(s) by which the WNK-SPAK/OSR1 pathway is regulated remain unclear. In this report, we identified WNK4 as an interacting partner of a recently identified MAP3K, apoptosis signal-regulating kinase 3 (ASK3). We found that WNK4 is phosphorylated in an ASK3 kinase activity-dependent manner. By exploring the ASK3-dependent phosphorylation sites, we identified Ser575 as a novel phosphorylation site in WNK4 by LC-MS/MS analysis. ASK3-dependent WNK4 Ser575 phosphorylation was mediated by the p38MAPK-MAPK-activated protein kinase (MK) pathway. Osmotic stress, as well as hypotonic low-chloride stimulation, increased WNK4 Ser575 phosphorylation via the p38MAPK-MK pathway. ASK3 was required for the p38MAPK activation induced by hypotonic stimulation but was not required for that induced by hypertonic stimulation or hypotonic low-chloride stimulation. Our results suggest that the p38MAPK-MK pathway might regulate WNK4 in an osmotic stress-dependent manner but its upstream regulators might be divergent depending on the types of osmotic stimuli.


Subject(s)
Osmotic Pressure , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing , Amino Acids/metabolism , Carrier Proteins/metabolism , Cell Line , Enzyme Activation , Gene Expression , Humans , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Microfilament Proteins , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics
6.
Cell Microbiol ; 18(2): 282-301, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26294282

ABSTRACT

Botulinum neurotoxins (BoNTs) are responsible for severe flaccid paralysis (botulism), which in most cases enter the organism via the digestive tract and then disseminate into the blood or lymph circulation to target autonomic and motor nerve endings. The passage way of BoNTs alone or in complex forms with associated nontoxic proteins through the epithelial barrier of the digestive tract still remains unclear. Here, we show using an in vivo model of mouse ligated intestinal loop that BoNT/B alone or the BoNT/B C-terminal domain of the heavy chain (HCcB), which interacts with cell surface receptors, translocates across the intestinal barrier. The BoNT/B or HCcB translocation through the intestinal barrier occurred via an endocytosis-dependent mechanism within 10-20 min, because Dynasore, a potent endocytosis inhibitor, significantly prevented BoNT/B as well as HCcB translocation. We also show that HCcB or BoNT/B specifically targets neuronal cells and neuronal extensions in the intestinal submucosa and musculosa expressing synaptotagmin, preferentially cholinergic neurons and to a lower extent other neuronal cell types, notably serotonergic neurons. Interestingly, rare intestinal epithelial cells accumulated HCcB suggesting that distinct cell types of the intestinal epithelium, still undefined, might mediate efficient translocation of BoNT/B.


Subject(s)
Botulinum Toxins, Type A/metabolism , Cholinergic Neurons/metabolism , Endocytosis , Intestinal Mucosa/metabolism , Animals , Epithelial Cells/metabolism , Mice , Protein Transport , Serotonergic Neurons/metabolism , Time Factors
7.
J Am Soc Nephrol ; 27(3): 835-46, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26293821

ABSTRACT

The iron-regulatory peptide hepcidin exhibits antimicrobial activity. Having previously shown hepcidin expression in the kidney, we addressed its role in urinary tract infection (UTI), which remains largely unknown. Experimental UTI was induced in wild-type (WT) and hepcidin-knockout (Hepc-/-) mice using the uropathogenic Escherichia coli CFT073 strain. Compared with infected WT mice, infected Hepc-/- mice showed a dramatic increase in renal bacterial load. Moreover, bacterial invasion was significantly dampened by the pretreatment of WT mice with hepcidin. Infected Hepc-/- mice exhibited decreased iron accumulation in the renal medulla and significant attenuation of the renal inflammatory response. Notably, we demonstrated in vitro bacteriostatic activity of hepcidin against CFT073. Furthermore, CFT073 repressed renal hepcidin, both in vivo and in cultured renal cells, and reduced phosphorylation of SMAD kinase in vivo, suggesting a bacterial strategy to escape the antimicrobial activities of hepcidin. In conclusion, we provide new mechanisms by which hepcidin contributes to renal host defense and suggest that targeting hepcidin offers a strategy to prevent bacterial invasion.


Subject(s)
Anti-Infective Agents/pharmacology , Escherichia coli Infections/metabolism , Escherichia coli/drug effects , Hepcidins/metabolism , Hepcidins/pharmacology , Urinary Tract Infections/metabolism , Animals , Anti-Infective Agents/metabolism , Bacterial Load/genetics , Cells, Cultured , Colony Count, Microbial , Cytokines/metabolism , Escherichia coli Infections/microbiology , Female , Hepcidins/genetics , Iron/metabolism , Kidney Medulla/cytology , Kidney Medulla/metabolism , Kidney Medulla/microbiology , Mice , Mice, Inbred CBA , Mice, Knockout , Nephritis/metabolism , Nephritis/microbiology , Nephritis/pathology , Neutrophils , Phosphorylation , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Smad Proteins/metabolism , Urinary Tract Infections/microbiology
8.
J Hypertens ; 33(8): 1709-15, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26132761

ABSTRACT

CASE REPORT: A 20-year-old woman presented with malignant hypertension associated with hypokalemia, metabolic alkalosis and elevated plasma renin and aldosterone levels. Computed tomography angiography (CTA) evidenced a 22 mm tissular mass in the posterior cortex of the left kidney, and 18F-flurodeoxyglucose PET (18-FDG PET) imaging showed no hypermetabolism of the tumour. Following nephron-sparing surgery, blood pressure and potassium levels rapidly normalized, allowing interruption of all treatments within 2 weeks. DISCUSSION: Reninoma is a rare juxtaglomerular cell tumour (JGCT) producing excessive amounts of renin resulting in severe hypertension. Pathological studies revealed that tumoural cells highly expressed renin and contained electron-dense structures, typical of renin-containing granules. Tumoural cells also exhibited the vascular cell surface marker CD34, but, in contrast with previous reports, did not express the tyrosine-protein kinase Kit (cKit or CD117). Dissociation of the tumour allowed to obtain confluent cultures of elongated smooth muscle actin (SMA)-positive cells producing high amounts of renin. However, after the first passage, subcultured human juxtaglomerular cells rapidly lost renin and CD34 expressions and their ability to produce renin. CONCLUSION: The present case of reninoma emphasizes the need for CTA in the etiologic work up of otherwise unexplained severe hypertension. 18-FDG PET imaging showed no hypermetabolism of the tumour, in accordance with its reported benignity. Pathological studies further emphasized that high expressions of renin and CD34 are typical hallmarks of reninoma. Although CD117 has been proposed to represent a reliable marker of JGCT, the present findings indicate that reninomas may not always express this marker.


Subject(s)
Juxtaglomerular Apparatus/diagnostic imaging , Juxtaglomerular Apparatus/pathology , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/pathology , Myocytes, Smooth Muscle/pathology , Renin/metabolism , Angiography , Female , Humans , Hypertension, Malignant/etiology , Kidney Neoplasms/metabolism , Renin/blood , Tomography, X-Ray Computed , Young Adult
9.
Cell Microbiol ; 16(10): 1503-17, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24779433

ABSTRACT

Uropathogenic Escherichia coli (UPEC) colonizing kidneys is the main cause of acute pyelonephritis. TLR5 that senses flagellin was shown to be highly expressed in the bladder and to participate in host defence against flagellated UPEC, although its role in kidneys still remains elusive. Here we show that TLR5 is expressed in renal medullary collecting duct (MCD) cells, which represent a preferential site of UPEC adhesion. Flagellin, like lipopolysaccharide, stimulated the production of the chemoattractant chemokines CXCL1 and CXCL2, and subsequent migration capacity of neutrophils in cultured wild-type (WT) and Tlr4(-/-) MCDs, but not in Tlr5(-/-) MCDs. UPEC can translocate across intact MCD layers without altering tight junctions. Strikingly, the invasion capacity and transcellular translocation of the UPEC strain HT7 were significantly lower in Tlr5(-/-) than in WT MCDs. The non-motile HT7ΔfliC mutant lacking flagellin also exhibited much lower translocation capacities than the HT7 isolates. Finally, Tlr5(-/-) kidneys exhibited less infiltrating neutrophils than WT kidneys one day after the transurethral inoculation of HT7, and greater delayed renal bacterial loads in the day 4 post-infected Tlr5(-/-) kidneys. Overall, these findings indicate that the epithelial TLR5 participates to renal antibacterial defence, but paradoxically favours the translocation of UPEC across intact MCD cell layers.


Subject(s)
Escherichia coli Infections/immunology , Flagellin/immunology , Kidney Tubules, Collecting/immunology , Toll-Like Receptor 5/immunology , Uropathogenic Escherichia coli/pathogenicity , Animals , Bacterial Adhesion/physiology , Bacterial Load/immunology , Chemokine CXCL1/biosynthesis , Chemokine CXCL2/biosynthesis , Escherichia coli Infections/microbiology , Female , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , Pyelonephritis/immunology , Pyelonephritis/microbiology , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 5/genetics , Urinary Bladder/immunology , Urinary Bladder/metabolism , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/immunology
10.
PLoS Negl Trop Dis ; 8(1): e2664, 2014.
Article in English | MEDLINE | ID: mdl-24498450

ABSTRACT

BACKGROUND: Leptospira (L.) interrogans are bacteria responsible for a worldwide reemerging zoonosis. Rodents carry L. interrogans asymptomatically in their kidneys and excrete bacteria in the urine, contaminating the environment. Humans get infected through skin contact and develop a mild or severe leptospirosis that may lead to renal failure and fibrosis. L. interrogans provoke an interstitial nephritis, but the induction of fibrosis caused by L. interrogans has not been studied in murine models. Innate immune receptors from the TLR and NLR families have recently been shown to play a role in the development and progression of tissue fibrosis in the lung, liver and kidneys under different pathophysiological situations. We recently showed that TLR2, TLR4, and NLRP3 receptors were crucial in the defense against leptospirosis. Moreover, infection of a human cell line with L. interrogans was shown to induce TLR2-dependent production of fibronectin, a component of the extracellular matrix. Therefore, we thought to assess the presence of renal fibrosis in L. interrogans infected mice and to analyze the contribution of some innate immune pathways in this process. METHODOLOGY/PRINCIPAL FINDINGS: Here, we characterized by immunohistochemical studies and quantitative real-time PCR, a model of Leptospira-infected C57BL/6J mice, with chronic carriage of L. interrogans inducing mild renal fibrosis. Using various strains of transgenic mice, we determined that the renal infiltrates of T cells and, unexpectedly, TLR and NLR receptors, are not required to generate Leptospira-induced renal fibrosis. We also show that the iNOS enzyme, known to play a role in Leptospira-induced interstitial nephritis, also plays a role in the induction of renal fibrosis. CONCLUSION/SIGNIFICANCE: To our knowledge, this work provides the first experimental murine model of sustained renal fibrosis induced by a chronic bacterial infection that may be peculiar, since it does not rely on TLR or NLR receptors. This model may prove useful to test future therapeutic strategies to combat Leptospira-induced renal lesions.


Subject(s)
Fibrosis/pathology , Kidney Diseases/pathology , Leptospira interrogans/immunology , Leptospirosis/pathology , Nitric Oxide Synthase Type II/metabolism , Signal Transduction , Animals , Carrier State/immunology , Carrier State/microbiology , Female , Fibrosis/immunology , Fibrosis/microbiology , Immunohistochemistry , Kidney/immunology , Kidney/microbiology , Kidney/pathology , Kidney Diseases/immunology , Kidney Diseases/microbiology , Leptospirosis/immunology , Leptospirosis/microbiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/metabolism , Toll-Like Receptors/metabolism
11.
Cell Commun Signal ; 12: 8, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24479879

ABSTRACT

The calcineurin/nuclear factor of activated T cells (NFATs) signaling pathway plays a central role in T cell mediated adaptive immune responses, but a number of recent studies demonstrated that calcineurin/NFAT signaling also plays a key role in the control of the innate immune response by myeloid cells. Calcineurin inhibitors, such as cyclosporine A (CsA) and tacrolimus (FK506), are commonly used in organ transplantation to prevent graft rejection and in a variety of immune diseases. These immunosuppressive drugs have adverse effects and significantly increase host's susceptibility towards bacterial or fungal infections. Recent studies highlighted the role of NFAT signaling in fungal infection and in the control of the pattern recognition receptor nucleotide-binding oligomerization domain-containing protein 1 (NOD1), which predominantly senses invasive Gram-negative bacteria and mediates neutrophil phagocytic functions. This review summarises some of the current knowledge concerning the role of NFAT signaling in the innate immune response and the recent advances on NFAT-dependent inhibition of NOD1-mediated innate immune response caused by CsA, which may contribute to sensitizing transplant recipients to bacterial infection.


Subject(s)
Calcineurin/metabolism , Immunity, Innate , NFATC Transcription Factors/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Phagocytosis , Signal Transduction , Animals , Bacterial Infections/immunology , Calcineurin/genetics , Humans , NFATC Transcription Factors/genetics , Nod1 Signaling Adaptor Protein/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
PLoS Pathog ; 9(1): e1003152, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23382681

ABSTRACT

Acute pyelonephritis (APN), which is mainly caused by uropathogenic Escherichia coli (UPEC), is the most common bacterial complication in renal transplant recipients receiving immunosuppressive treatment. However, it remains unclear how immunosuppressive drugs, such as the calcineurin inhibitor cyclosporine A (CsA), decrease renal resistance to UPEC. Here, we investigated the effects of CsA in host defense against UPEC in an experimental model of APN. We show that CsA-treated mice exhibit impaired production of the chemoattractant chemokines CXCL2 and CXCL1, decreased intrarenal recruitment of neutrophils, and greater susceptibility to UPEC than vehicle-treated mice. Strikingly, renal expression of Toll-like receptor 4 (Tlr4) and nucleotide-binding oligomerization domain 1 (Nod1), neutrophil migration capacity, and phagocytic killing of E. coli were significantly reduced in CsA-treated mice. CsA inhibited lipopolysaccharide (LPS)-induced, Tlr4-mediated production of CXCL2 by epithelial collecting duct cells. In addition, CsA markedly inhibited Nod1 expression in neutrophils, macrophages, and renal dendritic cells. CsA, acting through inhibition of the nuclear factor of activated T-cells (NFATs), also markedly downregulated Nod1 in neutrophils and macrophages. Silencing the NFATc1 isoform mRNA, similar to CsA, downregulated Nod1 expression in macrophages, and administration of the 11R-VIVIT peptide inhibitor of NFATs to mice also reduced neutrophil bacterial phagocytosis and renal resistance to UPEC. Conversely, synthetic Nod1 stimulating agonists given to CsA-treated mice significantly increased renal resistance to UPEC. Renal transplant recipients receiving CsA exhibited similar decrease in NOD1 expression and neutrophil phagocytosis of E. coli. The findings suggest that such mechanism of NFATc1-dependent inhibition of Nod1-mediated innate immune response together with the decrease in Tlr4-mediated production of chemoattractant chemokines caused by CsA may contribute to sensitizing kidney grafts to APN.


Subject(s)
Chemokines/metabolism , Cyclosporine/adverse effects , Immunosuppressive Agents/adverse effects , Kidney Transplantation , Kidney/drug effects , Nod1 Signaling Adaptor Protein/metabolism , Pyelonephritis/chemically induced , Acute Disease , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Movement/drug effects , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Disease Models, Animal , Female , Gene Silencing , Humans , Immunity, Innate , Kidney/metabolism , Kidney/pathology , Kidney Transplantation/immunology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/antagonists & inhibitors , Neutrophils/drug effects , Neutrophils/metabolism , Organ Culture Techniques , Phagocytosis/drug effects
13.
Nat Commun ; 3: 1285, 2012.
Article in English | MEDLINE | ID: mdl-23250415

ABSTRACT

Changes in the osmolality of body fluids pose a serious danger to cells and living organisms, which have developed cellular systems to sense and respond to osmotic stress and to maintain homoeostasis of body fluid. However, these processes are incompletely understood in mammals. Here we show that apoptosis signal-regulating kinase 3 (ASK3) is predominantly expressed in the kidney and alters its kinase activity bidirectionally in response to osmotic stress. We further demonstrate that ASK3 interacts with WNK1, mutation in which causes an inherited form of hypertension in humans. Knockdown of Ask3 by short interfering RNA enhances the activation of the WNK1-SPAK/OSR1 signalling pathway. Moreover, Ask3 knockout mice exhibit a hypertensive phenotype, in addition to hyperactivation of SPAK/OSR1 in renal tubules. Our results suggest that ASK3 is a unique bidirectional responder to osmotic stress and that it has a role in the control of blood pressure as an upstream suppressor of the WNK1-SPAK/OSR1 signalling pathway.


Subject(s)
Blood Pressure/physiology , Kidney/physiology , MAP Kinase Kinase Kinases/physiology , Osmotic Pressure/physiology , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Animals , HeLa Cells , Humans , Kidney Tubules/physiology , Male , Mice , Mice, Knockout , Minor Histocompatibility Antigens , WNK Lysine-Deficient Protein Kinase 1
14.
EMBO Mol Med ; 4(12): 1308-19, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143987

ABSTRACT

Intestinal ischemia/reperfusion (I/R) injury causes inflammation and tissue damage and is associated with high morbidity and mortality. Uncontrolled activation of the innate immune system through toll-like receptors (Tlr) plays a key role in I/R-mediated tissue damage but the underlying mechanisms have not been fully resolved. Here, we identify post-transcriptional upregulation of the essential Tlr signalling molecule interleukin 1 receptor-associated kinase (Irak) 1 as the causative mechanism for post-ischemic immune hyper-responsiveness of intestinal epithelial cells. Increased Irak1 protein levels enhanced epithelial ligand responsiveness, chemokine secretion, apoptosis and mucosal barrier disruption in an experimental intestinal I/R model using wild-type, Irak1(-/-) and Tlr4(-/-) mice and ischemic human intestinal tissue. Irak1 accumulation under hypoxic conditions was associated with reduced K48 ubiquitination and enhanced Senp1-mediated deSUMOylation of Irak1. Importantly, administration of microRNA (miR)-146a or induction of miR-146a by the phytochemical diindolylmethane controlled Irak1 upregulation and prevented immune hyper-responsiveness in mouse and human tissue. These findings indicate that Irak1 accumulation triggers I/R-induced epithelial immune hyper-responsiveness and suggest that the induction of miR-146a offers a promising strategy to prevent I/R tissue injury.


Subject(s)
Down-Regulation , Interleukin-1 Receptor-Associated Kinases/genetics , Intestine, Small/metabolism , Ischemia/genetics , MicroRNAs/metabolism , Reperfusion Injury/genetics , Animals , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Intestine, Small/blood supply , Intestine, Small/immunology , Ischemia/metabolism , Mice , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Signal Transduction
15.
PLoS One ; 7(9): e46547, 2012.
Article in English | MEDLINE | ID: mdl-23029548

ABSTRACT

Increasing numbers of pyelonephritis-associated uropathogenic Escherichia coli (UPEC) are exhibiting high resistance to antibiotic therapy. They include a particular clonal group, the CTX-M-15-producing O25b:H4-ST131 clone, which has been shown to have a high dissemination potential. Here we show that a representative isolate of this E. coli clone, referred to as TN03, has enhanced metabolic capacities, acts as a potent intestine- colonizing strain, and displays the typical features of UPEC strains. In a modified streptomycin-treated mouse model of intestinal colonization where streptomycin was stopped 5 days before inoculation, we show that TN03 outcompetes the commensal E. coli strains K-12 MG1655, IAI1, and ED1a at days 1 and 7. Using an experimental model of ascending UTI in C3H/HeN mice, we then show that TN03 colonized the urinary tract. One week after the transurethral inoculation of the TN03 isolates, the bacterial loads in the bladder and kidneys were significantly greater than those of two other UPEC strains (CFT073 and HT7) belonging to the same B2 phylogenetic group. The differences in bacterial loads did not seem to be directly linked to differences in the inflammatory response, since the intrarenal expression of chemokines and cytokines and the number of polymorphonuclear neutrophils attracted to the site of inflammation was the same in kidneys colonized by TN03, CFT073, or HT7. Lastly, we show that in vitro TN03 has a high maximum growth rate in both complex (Luria-Bertani and human urine) and minimum media. In conclusion, our findings indicate that TN03 is a potent UPEC strain that colonizes the intestinal tract and may persist in the kidneys of infected hosts.


Subject(s)
Escherichia coli Infections/microbiology , Escherichia coli Proteins/biosynthesis , Intestines/microbiology , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/enzymology , beta-Lactamases/biosynthesis , Animals , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression , Humans , Kidney/immunology , Kidney/metabolism , Kidney/microbiology , Kidney Diseases/immunology , Kidney Diseases/microbiology , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C3H , Uropathogenic Escherichia coli/growth & development , Uropathogenic Escherichia coli/pathogenicity , beta-Lactam Resistance
16.
J Immunol ; 188(6): 2805-14, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22323544

ABSTRACT

Leptospira interrogans is responsible for a zoonotic disease known to induce severe kidney dysfunction and inflammation. In this work, we demonstrate that L. interrogans induces NLRP3 inflammasome-dependent secretion of IL-1ß through the alteration of potassium transport in bone marrow-derived macrophages. Lysosome destabilization also contributed to the IL-1ß production upon stimulation with live, but not dead, bacteria. Using bone marrow-derived macrophages from various TLRs and nucleotide-binding oligomerization domain-deficient mice, we further determined that IL-1ß production was dependent on TLR2 and TLR4, suggesting a participation of the leptospiral LPS to this process. Hypokaliemia in leptospirosis has been linked to the presence of glycolipoprotein, a cell wall component of L. interrogans that is known to inhibit the expression and functions of the Na/K-ATPase pump. We show in this study that glycolipoprotein activates the inflammasome and synergizes with leptospiral LPS to produce IL-1ß, mimicking the effect of whole bacteria. These results were confirmed in vivo, as wild-type mice expressed more IL-1ß in the kidney than TLR2/4-deficient mice 3 d postinfection with L. interrogans. Collectively, these findings provide the first characterization, to our knowledge, of bacteria-induced activation of the NLRP3 inflammasome through the downregulation of a specific host potassium transporter.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Inflammasomes/metabolism , Leptospirosis/metabolism , Macrophages/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Bacterial Proteins/immunology , Blotting, Western , Carrier Proteins/immunology , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Inflammasomes/immunology , Interleukin-1beta/biosynthesis , Interleukin-1beta/immunology , Leptospira/immunology , Leptospira/metabolism , Leptospirosis/immunology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
17.
PLoS One ; 6(8): e24277, 2011.
Article in English | MEDLINE | ID: mdl-21909387

ABSTRACT

The NaCl cotransporter (NCC) is essential for sodium reabsorption at the distal convoluted tubules (DCT), and its phosphorylation increases its transport activity and apical membrane localization. Although insulin has been reported to increase sodium reabsorption in the kidney, the linkage between insulin and NCC phosphorylation has not yet been investigated. This study examined whether insulin regulates NCC phosphorylation. In cultured mpkDCT cells, insulin increased phosphorylation of STE20/SPS1-related proline-alanine-rich kinase (SPAK) and NCC in a dose-dependent manner. This insulin-induced phosphorylation of NCC was suppressed in WNK4 and SPAK knockdown cells. In addition, Ly294002, a PI3K inhibitor, decreased the insulin effect on SPAK and NCC phosphorylation, indicating that insulin induces phosphorylation of SPAK and NCC through PI3K and WNK4 in mpkDCT cells. Moreover, acute insulin administration to mice increased phosphorylation of oxidative stress-responsive kinase-1 (OSR1), SPAK and NCC in the kidney. Time-course experiments in mpkDCT cells and mice suggested that SPAK is upstream of NCC in this insulin-induced NCC phosphorylation mechanism, which was confirmed by the lack of insulin-induced NCC phosphorylation in SPAK knockout mice. Moreover, insulin administration to WNK4 hypomorphic mice did not increase phosphorylation of OSR1, SPAK and NCC in the kidney, suggesting that WNK4 is also involved in the insulin-induced OSR1, SPAK and NCC phosphorylation mechanism in vivo. The present results demonstrated that insulin is a potent regulator of NCC phosphorylation in the kidney, and that WNK4 and SPAK are involved in this mechanism of NCC phosphorylation by insulin.


Subject(s)
Insulin/pharmacology , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/metabolism , Sodium Chloride Symporters/metabolism , Animals , Cells, Cultured , Gene Knockdown Techniques , Insulin/administration & dosage , Kidney Tubules, Distal/enzymology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Sodium Chloride, Dietary/pharmacology , Time Factors
18.
Cell Microbiol ; 13(8): 1107-13, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21615666

ABSTRACT

Urinary tract infections (UTIs), which are mainly due to uropathogenic Escherichia coli (UPEC), occur via the retrograde ascent of the bacteria along the urinary tract system. The adhesion and invasion mechanisms of UPEC have been extensively studied in bladder epithelial cells, but less is known about the role of renal tubule epithelial cells (RTEC) in renal antibacterial defences. This review considers recent advances in the understanding of the role of RTECs in inducing an innate immune response mediated by Toll-like receptors (TLRs) in experimental UTI. Collecting duct cells are a preferential site of adhesion of UPEC colonizing the kidneys. Epithelial TLR4 activation induces an inflammatory response and the recruitment of lipid rafts to the plasma membrane, both of which facilitate the transcytosis of non-cytolytic UPEC strains across intact collecting duct cell layers to invade the renal interstitium. Arginine vasopressin, which regulates water absorption in the collecting duct, also acts as a potent modulator of the TLR4-mediated intrarenal innate response caused by UPEC. The role of epithelial TLR5 in renal host defences is also discussed. These findings highlight the role of RTECs in triggering the innate immune response in the context of ascending UTIs.


Subject(s)
Epithelial Cells/immunology , Escherichia coli Infections/immunology , Kidney Tubules, Collecting/immunology , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli/immunology , Arginine Vasopressin/metabolism , Bacterial Adhesion , Humans , Immunity, Innate , Models, Biological , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 5/metabolism , Transcytosis , Uropathogenic Escherichia coli/pathogenicity
19.
PLoS Pathog ; 6(11): e1001194, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-21124989

ABSTRACT

The epithelium efficiently attracts immune cells upon infection despite the low number of pathogenic microbes and moderate levels of secreted chemokines per cell. Here we examined whether horizontal intercellular communication between cells may contribute to a coordinated response of the epithelium. Listeria monocytogenes infection, transfection, and microinjection of individual cells within a polarized intestinal epithelial cell layer were performed and activation was determined at the single cell level by fluorescence microscopy and flow cytometry. Surprisingly, chemokine production after L. monocytogenes infection was primarily observed in non-infected epithelial cells despite invasion-dependent cell activation. Whereas horizontal communication was independent of gap junction formation, cytokine secretion, ion fluxes, or nitric oxide synthesis, NADPH oxidase (Nox) 4-dependent oxygen radical formation was required and sufficient to induce indirect epithelial cell activation. This is the first report to describe epithelial cell-cell communication in response to innate immune activation. Epithelial communication facilitates a coordinated infectious host defence at the very early stage of microbial infection.


Subject(s)
Cell Communication , Epithelial Cells/microbiology , Intestine, Small/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/immunology , Animals , Bacterial Toxins/metabolism , Blotting, Western , Cell Adhesion , Cells, Cultured , Cytokines/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Immunity, Innate , Immunoblotting , Immunoprecipitation , Intestine, Small/metabolism , Intestine, Small/microbiology , Listeriosis/microbiology , Listeriosis/pathology , Mice , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
20.
Kidney Int ; 78(7): 650-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20613715

ABSTRACT

Patients with proteinuric kidney diseases often have symptoms of salt and water retention. It has been hypothesized that dysregulated sodium absorption is due to increased proteolytic cleavage of epithelial sodium channels (ENaCs) and increased Na,K-ATPase expression. Microarray analysis identified a reduction in kidney corin mRNA expression in rat models of puromycin aminonucleoside-induced nephrotic syndrome and acute anti-Thy1 glomerulonephritis (GN). As atrial natriuretic peptide (ANP) resistance is a mechanism accounting for volume retention, we analyzed the renal expression and function of corin; a type II transmembrane serine protease that converts pro-ANP to active ANP. Immunohistochemical analysis found that corin colocalized with ANP. The nephrotic and glomerulonephritic models exhibited concomitant increased pro-ANP and decreased ANP protein levels in the kidney consistent with low amounts of corin. Importantly, kidneys from corin knockout mice had increased amounts of renal ß-ENaC and its activators, phosphodiesterase (PDE) 5 and protein kinase G II, when compared to wild-type mice. A similar expression profile was also found in cell culture suggesting the increase in PDE5 and kinase G II could account for the increase in ß-ENaC seen in nephrotic syndrome and GN. Thus, we suggest that corin might be involved in the salt retention seen in glomerular diseases.


Subject(s)
Kidney/metabolism , Proteinuria/metabolism , Serine Endopeptidases/physiology , Sodium/metabolism , Animals , Aquaporin 2/analysis , Atrial Natriuretic Factor/biosynthesis , Cyclic AMP/biosynthesis , Epithelial Sodium Channels/analysis , Glomerulonephritis/metabolism , HEK293 Cells , Humans , Male , Mice , Nephrotic Syndrome/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...