Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Microorganisms ; 11(8)2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37630625

ABSTRACT

Food is often contaminated with Escherichia coli (E. coli) bacteria strains, which have been associated with different diseases, including urinary tract infections. The consumption of meat by humans is a potential route of transmission of antimicrobial resistance, and food-producing animals have been associated as a major reservoir of resistant bacterial strains. The aim of this study was to determine the presence of the E. coli strains producing the CNF-1 toxin in pig kidneys. Pig kidneys were collected from a Mexican slaughterhouse and classified according to their coloration into reddish kidneys (RK) and yellowish kidneys (YK). A tissue sample from each kidney was processed for histological analysis, the presence of E. coli was determined by conventional PCR assay, and the CNF-1 toxin was detected by both conventional PCR and Western blotting. Herein, an inflammatory cell infiltrate was found in all collected kidneys, regardless of macroscopic differences. Surprisingly, E. coli and the CNF-1 toxin were detected in all kidney samples. We clearly demonstrate contamination by CNF-1 toxin-producing E. coli in pork kidneys from a slaughterhouse, even in those without apparent damage. This suggests that pork may serve as a reservoir for pathogens, representing an important risk to human health.

2.
Gut Microbes ; 15(1): 2229569, 2023.
Article in English | MEDLINE | ID: mdl-37417545

ABSTRACT

Colorectal cancer (CRC) patients are frequently colonized by colibactin-producing Escherichia coli (CoPEC) (>40%), which enhances tumorigenesis in mouse models of CRC. We observed that 50% of CoPEC also contains the cnf1 gene, which encodes cytotoxic necrotizing factor-1 (CNF1), an enhancer of the eukaryotic cell cycle. The impact of its co-occurrence with colibactin (Clb) has not yet been investigated. We evaluated the impact of CNF1 on colorectal tumorigenesis using human colonic epithelial HT-29 cells and CRC-susceptible ApcMin/+ mice inoculated with the CoPEC 21F8 clinical strain (Clb+Cnf+) or 21F8 isogenic mutants (Clb+Cnf-, Clb-Cnf+ and Clb-Cnf-). Infection with the Clb+Cnf- strain induced higher levels of inflammatory cytokines and senescence markers both in vitro and in vivo compared to those induced by infection with the Clb+Cnf+ strain. In contrast, the Clb+Cnf- and Clb+Cnf+ strains generated similar levels of DNA damage in HT-29 cells and in colonic murine tissues. Furthermore, the ApcMin/+ mice inoculated with the Clb+Cnf- strain developed significantly more tumors than the mice inoculated with the Clb+Cnf+ strain or the isogenic mutants, and the composition of their microbiota was changed. Finally, rectal administration of the CNF1 protein in ApcMin/+ mice inoculated with the Clb+Cnf- strain significantly decreased tumorigenesis and inflammation. Overall, this study provides evidence that CNF1 decreases the carcinogenic effects of CoPEC in ApcMin/+ mice by decreasing CoPEC-induced cellular senescence and inflammation.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Gastrointestinal Microbiome , Mice , Humans , Animals , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Colon , Carcinogenesis , Cell Transformation, Neoplastic , Inflammation
3.
J Cell Mol Med ; 27(10): 1315-1326, 2023 05.
Article in English | MEDLINE | ID: mdl-37078409

ABSTRACT

The bacterial product CNF1, through its action on the Rho GTPases, is emerging as a modulator of crucial signalling pathways involved in selected neurological diseases characterized by mitochondrial dysfunctions. Mitochondrial impairment has been hypothesized to have a key role in paramount mechanisms underlying Rett syndrome (RTT), a severe neurologic rare disorder. CNF1 has been already reported to have beneficial effects in mouse models of RTT. Using human RTT fibroblasts from four patients carrying different mutations, as a reliable disease-in-a-dish model, we explored the cellular and molecular mechanisms, which can underlie the CNF1-induced amelioration of RTT deficits. We found that CNF1 treatment modulates the Rho GTPases activity of RTT fibroblasts and induces a considerable re-organization of the actin cytoskeleton, mainly in stress fibres. Mitochondria of RTT fibroblasts show a hyperfused morphology and CNF1 decreases the mitochondrial mass leaving substantially unaltered the mitochondrial dynamic. From a functional perspective, CNF1 induces mitochondrial membrane potential depolarization and activation of AKT in RTT fibroblasts. Given that mitochondrial quality control is altered in RTT, our results are suggestive of a reactivation of the damaged mitochondria removal via mitophagy restoration. These effects can be at the basis of the beneficial effects of CNF1 in RTT.


Subject(s)
Escherichia coli Proteins , Rett Syndrome , Mice , Animals , Humans , Rett Syndrome/drug therapy , Rett Syndrome/genetics , Rett Syndrome/metabolism , rho GTP-Binding Proteins/metabolism , Pilot Projects , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/pharmacology , Mitochondria/metabolism , Fibroblasts/metabolism
4.
Gut Microbes ; 14(1): 2121577, 2022.
Article in English | MEDLINE | ID: mdl-36154446

ABSTRACT

Epidemiological projections point to acquisition of ever-expanding multidrug resistance (MDR) by Escherichia coli, a commensal of the digestive tract and a source of urinary tract pathogens. Bioinformatics analyses of a large collection of E. coli genomes from EnteroBase, enriched in clinical isolates of worldwide origins, suggest the Cytotoxic Necrotizing Factor 1 (CNF1)-toxin encoding gene, cnf1, is preferentially distributed in four common sequence types (ST) encompassing the pandemic E. coli MDR lineage ST131. This lineage is responsible for a majority of extraintestinal infections that escape first-line antibiotic treatment, with known enhanced capacities to colonize the gastrointestinal tract. Statistical projections based on this dataset point to a global expansion of cnf1-positive multidrug-resistant ST131 strains from subclade H30Rx/C2, accounting for a rising prevalence of cnf1-positive strains in ST131. Despite the absence of phylogeographical signals, cnf1-positive isolates segregated into clusters in the ST131-H30Rx/C2 phylogeny, sharing a similar profile of virulence factors and the same cnf1 allele. The suggested dominant expansion of cnf1-positive strains in ST131-H30Rx/C2 led us to uncover the competitive advantage conferred by cnf1 for gut colonization to the clinical strain EC131GY ST131-H30Rx/C2 versus cnf1-deleted isogenic strain. Complementation experiments showed that colon tissue invasion was compromised in the absence of deamidase activity on Rho GTPases by CNF1. Hence, gut colonization factor function of cnf1 was confirmed for another clinical strain ST131-H30Rx/C2. In addition, functional analysis of the cnf1-positive clinical strain EC131GY ST131-H30Rx/C2 and a cnf1-deleted isogenic strain showed no detectable impact of the CNF1 gene on bacterial fitness and inflammation during the acute phase of bladder monoinfection. Together these data argue for an absence of role of CNF1 in virulence during UTI, while enhancing gut colonization capacities of ST131-H30Rx/C2 and suggested expansion of cnf1-positive MDR isolates in subclade ST131-H30Rx/C2.


Subject(s)
Bacterial Toxins , Escherichia coli Infections , Escherichia coli Proteins , Gastrointestinal Microbiome , Anti-Bacterial Agents/pharmacology , Bacterial Toxins/genetics , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Humans , Virulence Factors/genetics , beta-Lactamases/genetics , beta-Lactamases/metabolism , rho GTP-Binding Proteins
5.
J Med Life ; 15(5): 650-654, 2022 May.
Article in English | MEDLINE | ID: mdl-35815089

ABSTRACT

Uropathogenic Escherichia coli (UPEC) harbors virulence factors responsible for bacterial adhesion and invasion. In addition, the bacterium is accountable for the occurrence of pediatric urinary tract infections globally and is becoming problematic due to the emergence of antimicrobial resistance. The current research investigated UPEC prevalence, virulence characteristics, and antimicrobial resistance in pediatric urinary tract infection (UTI). 200 urine specimens were taken from hospitalized pediatric patients who suffered from UTIs. E. coli was recovered from urine specimens using the microbial culture. Disc diffusion method was used to assess antimicrobial resistance and polymerase chain reaction (PCR) to assess the virulence factors distribution amongst the UPEC bacteria. Seventy-five out of 250 (30.00%) urine samples were positive for the UPEC bacteria. The UPEC prevalence amongst pediatric patients was 25.83% and 33.84%, respectively. UPEC bacteria harbored the maximum resistance toward gentamicin (45.33%), ampicillin (44.00%), and ciprofloxacin (40.00%). Cytotoxic necrotizing factor 1 (Cnf1) (53.33%) and pyelonephritis-associated pil (pap) (42.66%) were the most frequently identified virulence factors amongst the UPEC bacteria. The high prevalence of UPEC isolates harboring antimicrobial resistance and virulence factors suggest that diseases caused by them need more expansive healthcare monitoring with essential demand for novel antimicrobials.


Subject(s)
Escherichia coli Infections , Urinary Tract Infections , Uropathogenic Escherichia coli , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Child , Drug Resistance, Bacterial , Escherichia coli Infections/drug therapy , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Humans , Urinary Tract Infections/drug therapy , Virulence , Virulence Factors/genetics
6.
mBio ; 13(4): e0114722, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35766380

ABSTRACT

Urinary tract infections (UTIs) are a global public health concern, which is mainly caused by uropathogenic Escherichia coli (UPEC). Cytotoxic necrotizing factor 1 (CNF1) is a key UPEC toxin and regulates multiple host cellular processes through activating the Rho GTPases; however, the effect of CNF1 on macrophage polarization remains unknown. Here, we found that CNF1 promoted M1 macrophage polarization through regulating NF-κB and JAK-STAT1 signaling pathways in kidney at an early stage of acute UTIs. Notably, we identified CNF1 could directly interact with JAK1/2 through its domain without Rho GTPases activation, which induced JAK1/2 phosphorylation, subsequent STAT1 activation and M1 polarization. Moreover, CNF1 exhibited liquid-liquid phase separation (LLPS) to induce a CNF1-JAK1/2 complex, promoting macrophage reprogramming. These findings highlight the LLPS-dependent and Rho GTPase-independent effect of CNF1 as an adaptor on interfering with host cell signals. IMPORTANCE CNF1 is a key toxin secreted by UPEC, which induces inflammation during UPEC infections. CNF1 is well known to activate Rho GTPases to disturb host cell signaling pathways. Macrophage reprogramming plays important roles in inflammation; however, the effect of CNF1 on macrophage polarization is not reported. This study demonstrated the role and mechanism of CNF1 in promoting M1 macrophage polarization during UPEC-induced acute kidney infections. Importantly, we identified Rho GTPase-independent effect of CNF1 as an adaptor on interfering with host cell signals and demonstrated that CNF1 exhibited LLPS to drive its interaction with host proteins, which improve our understanding of the UPEC-host interactions and UTI pathogenesis.


Subject(s)
Bacterial Toxins , Escherichia coli Proteins , Urinary Tract Infections , Uropathogenic Escherichia coli , Bacterial Toxins/metabolism , Escherichia coli Proteins/metabolism , Humans , Inflammation , Janus Kinase 1/metabolism , Macrophages/metabolism , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/metabolism , rho GTP-Binding Proteins/metabolism
7.
Int J Mol Sci ; 23(10)2022 May 17.
Article in English | MEDLINE | ID: mdl-35628403

ABSTRACT

Simulated microgravity (SMG) inhibits osteoblast differentiation (OBD) and induces bone loss via the inhibition of the Wnt/ß-catenin pathway. However, the mechanism by which SMG alters the Wnt/ß-catenin pathway is unknown. We previously demonstrated that SMG altered the focal adhesion kinase (FAK)-regulated mTORC1, AMPK and ERK1/2 pathways, leading to the inhibition of tumor cell proliferation/metastasis and promoting cell apoptosis. To examine whether FAK similarly mediates SMG-dependent changes to Wnt/ß-catenin in osteoblasts, we characterized mouse MC3T3-E1 cells cultured under clinostat-modeled SMG (µg) conditions. Compared to cells cultured under ground (1 g) conditions, SMG reduces focal adhesions, alters cytoskeleton structures, and down-regulates FAK, Wnt/ß-catenin and Wnt/ß-catenin-regulated molecules. Consequently, protein-2 (BMP2), type-1 collagen (COL1), alkaline-phosphatase activity and matrix mineralization are all inhibited. In the mouse hindlimb unloading (HU) model, SMG-affected tibial trabecular bone loss is significantly reduced, according to histological and micro-computed tomography analyses. Interestingly, the FAK activator, cytotoxic necrotizing factor-1 (CNF1), significantly suppresses all of the SMG-induced alterations in MC3T3-E1 cells and the HU model. Therefore, our data demonstrate the critical role of FAK in the SMG-induced inhibition of OBD and bone loss via the Wnt/ß-catenin pathway, offering FAK signaling as a new therapeutic target not only for astronauts at risk of OBD inhibition and bone loss, but also osteoporotic patients.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases , Osteoblasts , Weightlessness , Wnt Signaling Pathway , beta Catenin , 3T3 Cells , Animals , Enzyme Activation , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , X-Ray Microtomography , beta Catenin/metabolism
8.
Am J Cancer Res ; 12(2): 651-660, 2022.
Article in English | MEDLINE | ID: mdl-35261793

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer death worldwide. The risk of developing CRC is influenced by both environmental and genetic factors. Recently, chronic inflammation and gut microbiota modifications have been associated with increased CRC risk. Escherichia coli belongs to the commensal intestinal flora and can become highly pathogenic following the acquisition of genes coding for virulence factors, such as the cytotoxic necrotizing factor type 1 (CNF1). Numerous reports highlight that, besides exerting direct effects on epithelial cells, CNF1 can also act on immune cells, modulating their responses and possibly contributing to disease development. In the present review, we summarized the key studies addressing the immunomodulatory functions of CNF1 and discussed the contribution that CNF1 can bring about to CRC through the creation of a pro-inflammatory microenvironment.

9.
Int J Mol Sci ; 22(18)2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34576292

ABSTRACT

The cytotoxic necrotizing factor 1 (CNF1) toxin from uropathogenic Escherichia coli constitutively activates Rho GTPases by catalyzing the deamidation of a critical glutamine residue located in the switch II (SWII). In crystallographic structures of the CNF1 catalytic domain (CNF1CD), surface-exposed P768 and P968 peptidyl-prolyl imide bonds (X-Pro) adopt an unusual cis conformation. Here, we show that mutation of each proline residue into glycine abrogates CNF1CD in vitro deamidase activity, while mutant forms of CNF1 remain functional on RhoA in cells. Using molecular dynamics simulations coupled to protein-peptide docking, we highlight the long-distance impact of peptidyl-prolyl cis-trans isomerization on the network of interactions between the loops bordering the entrance of the catalytic cleft. The energetically favorable isomerization of P768 compared with P968, induces an enlargement of loop L1 that fosters the invasion of CNF1CD catalytic cleft by a peptide encompassing SWII of RhoA. The connection of the P968 cis isomer to the catalytic cysteine C866 via a ladder of stacking interactions is alleviated along the cis-trans isomerization. Finally, the cis-trans conversion of P768 favors a switch of the thiol side chain of C866 from a resting to an active orientation. The long-distance impact of peptidyl-prolyl cis-trans isomerizations is expected to have implications for target modification.


Subject(s)
Bacterial Toxins/chemistry , Catalytic Domain , Escherichia coli Proteins/chemistry , Molecular Dynamics Simulation , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Isomerism , Molecular Docking Simulation , Protein Binding , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/metabolism
10.
Gut Pathog ; 11: 22, 2019.
Article in English | MEDLINE | ID: mdl-31139264

ABSTRACT

BACKGROUND: The colibactin and cytotoxic necrotizing factor 1 (Cnf 1) are toxins with cell cycle modulating effects that contribute to tumorgenesis and hyperproliferation. This study aimed to investigate the prevalence and pathologic effects of Cnf 1 and colibactin among hemolytic uropathogenic Escherichia coli (UPEC). The bioinformatics approach incorporated in this study aimed to expand the domain of the in vitro study and explore the prevalence of both toxins among other bacterial species. A total of 125 E. coli isolates were recovered from UTIs patients. The isolates were tested for their hemolytic activity, subjected to tissue culture and PCR assays to detect the phenotypic and genotypic features of both toxins. A rat ascending UTI in vivo model was conducted using isolates expressing or non-expressing Cnf 1 and colibactin (ClbA and ClbQ). The bioinformatics analyses were inferred by Maximum likelihood method and the evolutionary relatedness was deduced by MEGA X. RESULTS: Only 21 (16.8%) out of 125 isolates were hemolytic and 10 of these (47.62%) harbored the toxins encoding genes (cnf 1 +, clbA + and clbQ +). The phenotypic features of both toxins were exhibited by only 7 of the (cnf 1 + clbA + clbQ +) harboring isolates. The severest infections, hyperplastic and genotoxic changes in kidneys and bladders were observed in rats infected with the cnf 1 + clbA + clbQ + isolates. CONCLUSION: Only 33.3% of the hemolytic UPEC isolates exhibited the phenotypic and genotypic features of Cnf 1 and Colibactin. The in vivo animal model results gives an evidence of active Cnf 1 and Colibactin expression and indicates the risks associated with recurrent and chronic UTIs caused by UPEC. The bioinformatics analyses confirmed the predominance of colibactin pks island among Enterobacteriaceae family (92.86%), with the highest occurrence among Escherichia species (53.57%), followed by Klebsiella (28.57%), Citrobacter (7.14%), and Enterobacter species (3.57%). The Cnf 1 is predominant among Escherichia coli (94.05%) and sporadically found among Shigella species (1.08%), Salmonella enterica (0.54%), Yersinia pseudotuberculosis (1.08%), Photobacterium (1.08%), Moritella viscosa (0.54%), and Carnobacterium maltaromaticum (0.54%). A close relatedness was observed between the 54-kb pks island of Escherichia coli, the probiotic Escherichia coli Nissle 1917, Klebsiella aerogenes, Klebsiella pneumoniae and Citrobacter koseri.

11.
Methods Mol Biol ; 1821: 141-154, 2018.
Article in English | MEDLINE | ID: mdl-30062410

ABSTRACT

Bacterial pathogens developed several strategies to overcome defense systems of eukaryotic hosts. Within the infection process they need to attach to and cross through epithelial layers, escape from the innate and adaptive immune response, and find a physiological niche to survive. For this purpose bacteria developed toxins that specifically target central eukaryotic proteins, for example actin or Rho GTPases as regulators of the actin cytoskeleton. Some bacterial toxins catalyze a covalent modification of Rho GTPases to keep these molecular switches in a constitutive active or inactive state. This leads to rearrangement of the actin cytoskeleton. Toxin-treated cells show typical morphological changes depending on substrate specificity and action of the toxins. In this chapter I describe methods to illustrate how bacterial toxins may help to study the involvement of Rho GTPases in physiological and pathophysiological processes.


Subject(s)
Actin Cytoskeleton/metabolism , Bacterial Toxins/pharmacology , Protein Processing, Post-Translational/drug effects , rho GTP-Binding Proteins/metabolism , Actin Cytoskeleton/chemistry , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , HeLa Cells , Humans , rho GTP-Binding Proteins/chemistry
12.
Int J Mol Sci ; 19(6)2018 May 31.
Article in English | MEDLINE | ID: mdl-29857515

ABSTRACT

Pathogenic bacteria produce toxins to promote host invasion and, therefore, their survival. The extreme potency and specificity of these toxins confer to this category of proteins an exceptionally strong potential for therapeutic exploitation. In this review, we deal with cytotoxic necrotizing factor (CNF1), a cytotoxin produced by Escherichia coli affecting fundamental cellular processes, including cytoskeletal dynamics, cell cycle progression, transcriptional regulation, cell survival and migration. First, we provide an overview of the mechanisms of action of CNF1 in target cells. Next, we focus on the potential use of CNF1 as a pharmacological treatment in central nervous system's diseases. CNF1 appears to impact neuronal morphology, physiology, and plasticity and displays an antineoplastic activity on brain tumors. The ability to preserve neural functionality and, at the same time, to trigger senescence and death of proliferating glioma cells, makes CNF1 an encouraging new strategy for the treatment of brain tumors.


Subject(s)
Bacterial Toxins/pharmacology , Bacterial Toxins/therapeutic use , Brain Diseases/drug therapy , Brain Diseases/etiology , Molecular Targeted Therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bacterial Toxins/chemistry , Brain Diseases/metabolism , Brain Diseases/pathology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/pharmacology , Escherichia coli Proteins/therapeutic use , Gene Expression Regulation/drug effects , Humans , Neurons/drug effects , Neurons/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
13.
Int J Mol Sci ; 19(5)2018 May 08.
Article in English | MEDLINE | ID: mdl-29738516

ABSTRACT

Cytotoxic necrotizing factor 1 (CNF1) is a bacterial protein toxin primarily expressed by pathogenic Escherichia coli strains, causing extraintestinal infections. The toxin is believed to enhance the invasiveness of E. coli by modulating the activity of Rho GTPases in host cells, but it has interestingly also been shown to promote inflammation, stimulate host immunity and function as a potent immunoadjuvant. The mechanisms underlying the immunostimulatory properties of CNF1 are, however, poorly characterized, and little is known about the direct effects of the toxin on immune cells. Here, we show that CNF1 induces expression of maturation markers on human immature monocyte-derived dendritic cells (moDCs) without compromising cell viability. Consistent with the phenotypic maturation, CNF1 further triggered secretion of proinflammatory cytokines and increased the capacity of moDCs to stimulate proliferation of allogenic naïve CD4+ T cells. A catalytically inactive form of the toxin did not induce moDC maturation, indicating that the enzymatic activity of CNF1 triggers immature moDCs to undergo phenotypic and functional maturation. As the maturation of dendritic cells plays a central role in initiating inflammation and activating the adaptive immune response, the present findings shed new light on the immunostimulatory properties of CNF1 and may explain why the toxin functions as an immunoadjuvant.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bacterial Toxins/chemistry , Dendritic Cells/drug effects , Escherichia coli Proteins/chemistry , Inflammation/drug therapy , Adjuvants, Immunologic/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/pharmacology , Cell Survival/drug effects , Dendritic Cells/immunology , Escherichia coli/chemistry , Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , Escherichia coli Proteins/pharmacology , Humans , Inflammation/immunology , Inflammation/pathology , Monocytes/drug effects , Monocytes/immunology , rho GTP-Binding Proteins/genetics
14.
ChemMedChem ; 13(7): 754-761, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29359495

ABSTRACT

Cytotoxic necrotizing factor 1 (CNF1) is a toxin produced by pathogenic strains of Escherichia coli responsible for extra-intestinal infections. CNF1 deamidates Rac1, thereby triggering its permanent activation and worsening inflammatory reactions. Activated Rac1 is prone to proteasomal degradation. There is no targeted therapy against CNF1, despite its clinical relevance. In this work we developed a fluorescent cell-based immunoassay to screen for inhibitors of CNF1-induced Rac1 degradation among 1120 mostly approved drugs. Eleven compounds were found to prevent CNF1-induced Rac1 degradation, and five also showed a protective effect against CNF1-induced multinucleation. Finally, lasalocid, monensin, bepridil, and amodiaquine protected cells from both diphtheria toxin and CNF1 challenges. These data highlight the potential for drug repurposing to fight several bacterial infections and Rac1-based diseases.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Escherichia coli Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , rac1 GTP-Binding Protein/metabolism , Amodiaquine/pharmacology , Bacterial Toxins/adverse effects , Bacterial Toxins/metabolism , Bepridil/pharmacology , Diphtheria Toxin/adverse effects , Drug Repositioning , Escherichia coli/chemistry , Escherichia coli Proteins/adverse effects , Escherichia coli Proteins/metabolism , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunoassay , Lasalocid/pharmacology , Monensin/pharmacology , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/immunology
15.
Biotechnol Prog ; 34(1): 150-159, 2018 01.
Article in English | MEDLINE | ID: mdl-29063721

ABSTRACT

The Cytotoxic Necrotizing Factor 1 (CNF1) is a bacterial toxin secreted by certain Escherichia coli strains causing severe pathologies, making it a protein of pivotal interest in toxicology. In parallel, the CNF1 capability to influence important neuronal processes, like neuronal arborization, astrocytic support, and efficient ATP production, has been efficiently used in the treatment of neurological diseases, making it a promising candidate for therapy. Nonetheless, there are still some unsolved issues about the CNF1 mechanism of action and structuration probably caused by the difficulty to achieve sufficient amounts of the full-length protein for further studies. Here, we propose an efficient strategy for the production and purification of this toxin as a his-tagged recombinant protein from E. coli extracts (CNF1-H8). CNF1-H8 was expressed at the low temperature of 15°C to diminish its characteristic degradation. Then, its purification was achieved using an immobilized metal affinity chromatography (IMAC) and a size exclusion chromatography so as to collect up to 8 mg of protein per liter of culture in a highly pure form. Routine dynamic light scattering (DLS) experiments showed that the recombinant protein preparations were homogeneous and preserved this state for a long time. Furthermore, CNF1-H8 functionality was confirmed by testing its activity on purified RhoA and on HEp-2 cultured cells. Finally, a first structural characterization of the full-length toxin in terms of secondary structure and thermal stability was performed by circular dichroism (CD). These studies demonstrate that our system can be used to produce high quantities of pure recombinant protein for a detailed structural analysis. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 34:150-159, 2018.


Subject(s)
Bacterial Toxins/isolation & purification , Escherichia coli Proteins/isolation & purification , Escherichia coli/chemistry , Recombinant Proteins/isolation & purification , Bacterial Toxins/chemistry , Cell Line , Chromatography, Affinity , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Humans , Recombinant Proteins/chemistry , rhoA GTP-Binding Protein
16.
EMBO Rep ; 18(11): 1978-1990, 2017 11.
Article in English | MEDLINE | ID: mdl-28912123

ABSTRACT

Most type VI secretion systems (T6SSs) described to date are protein delivery apparatuses that mediate bactericidal activities. Several T6SSs were also reported to mediate virulence activities, although only few anti-eukaryotic effectors have been described. Here, we identify three T6SSs in the marine bacterium Vibrio proteolyticus and show that T6SS1 mediates bactericidal activities under warm marine-like conditions. Using comparative proteomics, we find nine potential T6SS1 effectors, five of which belong to the polymorphic MIX-effector class. Remarkably, in addition to six predicted bactericidal effectors, the T6SS1 secretome includes three putative anti-eukaryotic effectors. One of these is a MIX-effector containing a cytotoxic necrotizing factor 1 domain. We demonstrate that T6SS1 can use this MIX-effector to target phagocytic cells, resulting in morphological changes and actin cytoskeleton rearrangements. In conclusion, the V. proteolyticus T6SS1, a system homologous to one found in pathogenic vibrios, uses a suite of polymorphic effectors that target both bacteria and eukaryotic neighbors.


Subject(s)
Bacterial Proteins/genetics , Bacterial Toxins/genetics , Chromosomes, Bacterial/chemistry , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Type VI Secretion Systems/genetics , Vibrio/genetics , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/toxicity , Aquatic Organisms , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Chromosome Mapping , Coculture Techniques , Escherichia coli/drug effects , Escherichia coli/growth & development , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/toxicity , Mice , Phagocytes/cytology , Phagocytes/drug effects , Protein Domains , RAW 264.7 Cells , Type VI Secretion Systems/chemistry , Type VI Secretion Systems/metabolism , Vibrio/metabolism , Vibrio/pathogenicity , Virulence
17.
Oncotarget ; 8(14): 23539-23550, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28212563

ABSTRACT

Glioblastoma (GBM) is the most aggressive type of brain tumor. In this context, animal models represent excellent tools for the early detection and longitudinal mapping of neuronal dysfunction, that are critical in the preclinical validation of new therapeutic strategies. In a mouse glioma model, we developed sensitive behavioral readouts that allow early recognizing and following neurological symptoms. We injected GL261 cells into the primary motor cortex of syngenic mice and we used a battery of behavioral tests to longitudinally monitor the dysfunction induced by tumor growth. Grip strength test revealed an early onset of functional deficit associated to the glioma growth, with a significant forelimb weakness appearing 9 days after tumor inoculation. A later deficit was observed in the rotarod and in the grid walk tasks. Using this model, we found reduced tumor growth and maintenance of behavioral functions following treatment with Cytotoxic Necrotizing Factor 1 (CNF1) at a symptomatic stage. Our data provide a detailed and precise examination of how tumor growth reverberates on the behavioral functions of glioma-bearing mice, providing normative data for the study of therapeutic strategies for glioma treatment. The reduced tumor volume and robust functional sparing observed in CNF1-treated, glioma-bearing mice strengthen the notion that CNF1 delivery is a promising strategy for glioma therapy.


Subject(s)
Bacterial Toxins/pharmacology , Brain Neoplasms/drug therapy , Escherichia coli Proteins/pharmacology , Glioma/drug therapy , Motor Disorders/drug therapy , Analysis of Variance , Animals , Blotting, Western , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Disks Large Homolog 4 Protein , Glioma/pathology , Glioma/physiopathology , Guanylate Kinases/metabolism , Humans , Membrane Proteins/metabolism , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Disorders/physiopathology , Time Factors , Tumor Burden/drug effects
19.
Virulence ; 7(7): 806-18, 2016 10 02.
Article in English | MEDLINE | ID: mdl-27221788

ABSTRACT

Macrophages are a permissive niche for E. coli K1 multiplication for which the interaction of the bacterial outer membrane protein A and its cognate receptor CD64 are critical. Using in vitro immunofluorescence and live microscopy with ex vivo macrophage cultures from RFP-Lifeact mice, we show that cytotoxic necrotizing factor 1 (CNF1) secreted by E. coli K1 sequesters cellular actin toward microspike formation, thereby limiting actin availability for OmpA-mediated bacterial invasion. Surprisingly, the observed effects of CNF1 occur despite the absence of 67-kDa laminin receptor in macrophages. Concomitantly, the CNF1 deletion mutant of E. coli K1 (Δcnf1) invades macrophages and the brains of newborn mice in greater numbers compared to wild-type. However, the Δcnf1 strain induces less severe pathology in the brain. These results suggest a novel role for CNF1 in limiting E. coli K1 entry into macrophages while exacerbating disease severity in the brains of newborn mice.


Subject(s)
Bacterial Toxins/genetics , Escherichia coli Proteins/genetics , Escherichia coli/physiology , Macrophages/microbiology , Meningitis, Escherichia coli/microbiology , Actins , Animals , Animals, Newborn , Bacterial Outer Membrane Proteins/metabolism , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Brain/microbiology , Brain/pathology , Cells, Cultured , Disease Progression , Escherichia coli/genetics , Escherichia coli Proteins/immunology , Escherichia coli Proteins/metabolism , Fluorescent Antibody Technique , Humans , Macrophages/immunology , Meningitis, Escherichia coli/immunology , Mice , Receptors, IgG/metabolism , Sequence Deletion
20.
Emerg Infect Dis ; 22(3): 503-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26889961

ABSTRACT

We used multivirulence locus sequence typing to analyze 68 Yersinia pseudotuberculosis isolates from patients in Russia during 1973-2014, including 41 isolates from patients with Far East scarlet-like fever. Four genotypes were found responsible, with 1 being especially prevalent. Evolutionary analysis suggests that epidemiologic advantages could cause this genotype's dominance.


Subject(s)
Fever/epidemiology , Fever/microbiology , Genotype , Yersinia pseudotuberculosis Infections/epidemiology , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis/classification , Yersinia pseudotuberculosis/genetics , Animals , Bacterial Proteins/genetics , Evolution, Molecular , Genes, Essential , Humans , Multilocus Sequence Typing , Phylogeny , Polymorphism, Genetic , Russia/epidemiology , Serogroup , Virulence Factors/genetics , Yersinia pseudotuberculosis/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL