Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.911
1.
Cell Biochem Funct ; 42(4): e4026, 2024 Jun.
Article En | MEDLINE | ID: mdl-38693631

This work investigates the efficiency of cholecalciferol and low dose gamma radiation in modulating cytokine storm through their impact on inflammatory and anti-inflammatory cytokine and protecting against lung and liver injuries. Male Swiss albino mice were exposed to 0.2 Gy gamma radiation/week for four consecutive weeks then injected intraperitoneally (i.p) with a single dose of 8.3 × 106 CFU Escherichia coli/g b.w. then injected i.p. with 1.0 mg/kg cholecalciferol (Vit D3) for 7 days starting 4 h after E. coli injection. The results revealed that Cholecalciferol and low dose gamma radiation caused significant depletion in the severity of E. coli infection (colony forming unit per milliliter), log10 of E. coli, Tumor necrosis factor alpha, Interleukin 6, VEGF, alanine aminotransferase, and aspartate aminotransferase levels and significant elevation in IL-10, IL-4, and HO-1. Immunohistochemical analysis of caspase-3 expression in lung tissue section showed low caspase-3 expression in cholecalciferol and low dose gamma radiation treated group. Histopathological examinations were performed in both lung and liver tissues which also emphasis the biochemical findings. Our results exhibit the importance of cholecalciferol and low dose gamma radiation in improving liver function and providing anti-inflammatory response in diseases causing cytokine storm.


Cholecalciferol , Escherichia coli Infections , Escherichia coli , Gamma Rays , Animals , Mice , Cholecalciferol/pharmacology , Male , Escherichia coli Infections/drug therapy , Escherichia coli Infections/pathology , Liver/pathology , Liver/drug effects , Liver/metabolism , Lung/pathology , Lung/metabolism , Cytokines/metabolism , Cytokine Release Syndrome/pathology , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/etiology , Aspartate Aminotransferases/blood
2.
Cell Rep ; 43(4): 114051, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38564334

Uropathogenic Escherichia coli (UPEC) is the most common causative agent of urinary tract infection (UTI). UPEC invades bladder epithelial cells (BECs) via fusiform vesicles, escapes into the cytosol, and establishes biofilm-like intracellular bacterial communities (IBCs). Nucleoside-diphosphate kinase (NDK) is secreted by pathogenic bacteria to enhance virulence. However, whether NDK is involved in UPEC pathogenesis remains unclear. Here, we find that the lack of ndk impairs the colonization of UPEC CFT073 in mouse bladders and kidneys owing to the impaired ability of UPEC to form IBCs. Furthermore, we demonstrate that NDK inhibits caspase-1-dependent pyroptosis by consuming extracellular ATP, preventing superficial BEC exfoliation, and promoting IBC formation. UPEC utilizes the reactive oxygen species (ROS) sensor OxyR to indirectly activate the regulator integration host factor, which then directly activates ndk expression in response to intracellular ROS. Here, we reveal a signaling transduction pathway that UPEC employs to inhibit superficial BEC exfoliation, thus facilitating acute UTI.


Caspase 1 , Escherichia coli Infections , Nucleoside-Diphosphate Kinase , Pyroptosis , Urinary Tract Infections , Uropathogenic Escherichia coli , Uropathogenic Escherichia coli/pathogenicity , Animals , Urinary Tract Infections/microbiology , Urinary Tract Infections/pathology , Mice , Caspase 1/metabolism , Nucleoside-Diphosphate Kinase/metabolism , Nucleoside-Diphosphate Kinase/genetics , Escherichia coli Infections/microbiology , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL , Humans , Female , Urinary Bladder/microbiology , Urinary Bladder/pathology , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Signal Transduction
3.
Am J Physiol Cell Physiol ; 326(5): C1451-C1461, 2024 May 01.
Article En | MEDLINE | ID: mdl-38525539

Acute pyelonephritis (APN) is most frequently caused by uropathogenic Escherichia coli (UPEC), which ascends from the bladder to the kidneys during a urinary tract infection. Patients with APN have been reported to have reduced renal concentration capacity under challenged conditions, polyuria, and increased aquaporin-2 (AQP2) excretion in the urine. We have recently shown increased AQP2 accumulation in the plasma membrane in cell cultures exposed to E. coli lysates and in the apical plasma membrane of inner medullary collecting ducts in a 5-day APN mouse model. This study aimed to investigate if AQP2 expression in host cells increases UPEC infection efficiency and to identify specific bacterial components that mediate AQP2 plasma membrane insertion. As the transepithelial water permeability in the collecting duct is codetermined by AQP3 and AQP4, we also investigated whether AQP3 and AQP4 localization is altered in the APN mouse model. We show that AQP2 expression does not increase UPEC infection efficiency and that AQP2 was targeted to the plasma membrane in AQP2-expressing cells in response to the two pathogen-associated molecular patterns (PAMPs), lipopolysaccharide and peptidoglycan. In contrast to AQP2, the subcellular localizations of AQP1, AQP3, and AQP4 were unaffected both in lysate-incubated cell cultures and in the APN mouse model. Our finding demonstrated that cellular exposure to lipopolysaccharide and peptidoglycan can trigger the insertion of AQP2 in the plasma membrane revealing a new regulatory pathway for AQP2 plasma membrane translocation, which may potentially be exploited in intervention strategies.NEW & NOTEWORTHY Acute pyelonephritis (APN) is associated with reduced renal concentration capacity and increased aquaporin-2 (AQP2) excretion. Uropathogenic Escherichia coli (UPEC) mediates changes in the subcellular localization of AQP2 and we show that in vitro, these changes could be elicited by two pathogen-associated molecular patterns (PAMPs), namely, lipopolysaccharide and peptidoglycan. UPEC infection was unaltered by AQP2 expression and the other renal AQPs (AQP1, AQP3, and AQP4) were unaltered in APN.


Aquaporin 2 , Aquaporin 3 , Pyelonephritis , Uropathogenic Escherichia coli , Pyelonephritis/metabolism , Pyelonephritis/microbiology , Pyelonephritis/pathology , Animals , Aquaporin 2/metabolism , Mice , Uropathogenic Escherichia coli/metabolism , Aquaporin 3/metabolism , Aquaporin 3/genetics , Acute Disease , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Lipopolysaccharides/toxicity , Lipopolysaccharides/pharmacology , Cell Membrane/metabolism , Humans , Aquaporin 4/metabolism , Aquaporin 4/genetics , Peptidoglycan/metabolism , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , Disease Models, Animal
4.
Cell ; 187(8): 1874-1888.e14, 2024 Apr 11.
Article En | MEDLINE | ID: mdl-38518773

Infections of the lung cause observable sickness thought to be secondary to inflammation. Signs of sickness are crucial to alert others via behavioral-immune responses to limit contact with contagious individuals. Gram-negative bacteria produce exopolysaccharide (EPS) that provides microbial protection; however, the impact of EPS on sickness remains uncertain. Using genome-engineered Pseudomonas aeruginosa (P. aeruginosa) strains, we compared EPS-producers versus non-producers and a virulent Escherichia coli (E. coli) lung infection model in male and female mice. EPS-negative P. aeruginosa and virulent E. coli infection caused severe sickness, behavioral alterations, inflammation, and hypothermia mediated by TLR4 detection of the exposed lipopolysaccharide (LPS) in lung TRPV1+ sensory neurons. However, inflammation did not account for sickness. Stimulation of lung nociceptors induced acute stress responses in the paraventricular hypothalamic nuclei by activating corticotropin-releasing hormone neurons responsible for sickness behavior and hypothermia. Thus, EPS-producing biofilm pathogens evade initiating a lung-brain sensory neuronal response that results in sickness.


Escherichia coli Infections , Escherichia coli , Lung , Polysaccharides, Bacterial , Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Female , Male , Mice , Biofilms , Escherichia coli/physiology , Hypothermia/metabolism , Hypothermia/pathology , Inflammation/metabolism , Inflammation/pathology , Lung/microbiology , Lung/pathology , Pneumonia/microbiology , Pneumonia/pathology , Pseudomonas aeruginosa/physiology , Sensory Receptor Cells , Polysaccharides, Bacterial/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Nociceptors/metabolism
5.
J Ethnopharmacol ; 328: 118056, 2024 Jun 28.
Article En | MEDLINE | ID: mdl-38490287

ETHNOPHARMACOLOGICAL RELEVANCE: Urinary tract infections (UTIs) are globally prevalent infectious diseases, predominantly caused by uropathogenic Escherichia coli (UPEC). The misuse of antibiotics has led to the emergence of several drug-resistant strains. Traditional Chinese Medicine (TCM) has its own advantages in the treatment of UTIs. HJ granules is a herbal formula used for the treatment of UTIs. However, its mechanism of action is not clear. AIM OF THE STUDY: The aim of this study was to investigate the therapeutic efficacy and mechanism of action of HJ granules in a rat model of UTI caused by Escherichia coli (E coli) CFT073. MATERIALS AND METHODS: SD rats were selected to establish a rat UTI model by injecting UPEC strain CFT073 into the bladder using the transurethral placement method. HJ granules were administered to rats after modelling and the efficacy of HJ granule was investigated by measuring urinary decanalogue, inflammatory factors in bladder tissue and pathological changes in the bladder after 3d of administration. Expression of sonic hedgehog (SHH), NOD-like receptor thermoprotein domain 3 (NLRP3), apoptosis-associated speck-like protein (ASC) and activation of cysteinyl aspartate specific proteinase-1 (caspase-1) were detected by western blotting and immunofluorescence staining in rat bladder tissue. NLRP3, ASC and caspase-1, a cysteine-containing aspartic protein, were expressed and activated. RESULTS: The results showed that infection of rats with UPEC resulted in increased pH and erythrocytes in bladder irrigation fluid; increased expression of IL-1ß, IL-6 and SHH and decreased expression of IL-10 in bladder tissue; and significant upregulation of the expression of both SHH and NLRP3 inflammasom and significant activation of NLRP3 inflammasom. HJ granules significantly increased the concentration of IL-10 in the bladder, inhibited the expression of SHH and NLRP3 inflammasom in bladder tissue, and suppressed the activation of NLRP3 inflammasom, thereby reducing inflammatory lesions in bladder tissue. CONCLUSION: HJ granules may improve bladder injury and treat UTIs by inhibiting the expression and activation of NLRP3 inflammasom.


Escherichia coli Infections , Urinary Tract Infections , Uropathogenic Escherichia coli , Rats , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Escherichia coli , Interleukin-10 , Hedgehog Proteins , Escherichia coli Infections/drug therapy , Escherichia coli Infections/pathology , Rats, Sprague-Dawley , Urinary Tract Infections/drug therapy , Urinary Tract Infections/pathology , Caspase 1/metabolism
6.
Crit Rev Eukaryot Gene Expr ; 34(3): 83-99, 2024.
Article En | MEDLINE | ID: mdl-38305291

In Crohn's disease (CD), gut dysbiosis is marked by the prevalence of pathogenic bacterial species. Although several microbes have been reported as risk factors or causative agents of CD, it is not yet clear which is the real trigger of the disease. Thirty years ago, a new pathovar of Escherichia coli strain was isolated in the ileal mucosa of CD patients. This strain, called adherent invasive E. coli (AIEC), for its ability to invade the intestinal mucosa, could represent the causative agent of the disease. Several authors studied the mechanisms by which the AIEC penetrate and replicate within macrophages, and release inflammatory cytokines sustaining inflammation. In this review we will discuss about the role of AIEC in the pathogenesis of CD, the virulence factors mediating adhesion and invasion of AIEC in mucosal tissue, the environmental conditions improving AIEC survival and replication within macrophages. Finally, we will also give an overview of the new strategies developed to limit AIEC overgrowth.


Crohn Disease , Escherichia coli Infections , Humans , Crohn Disease/epidemiology , Crohn Disease/microbiology , Crohn Disease/pathology , Escherichia coli , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Bacterial Adhesion , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology
7.
Int Immunopharmacol ; 127: 111386, 2024 Jan 25.
Article En | MEDLINE | ID: mdl-38109839

Pathogenic Escherichia coli (E. coli) can cause intestinal diseases in humans and livestock, damage the intestinal barrier, increase systemic inflammation, and seriously threaten human health and the development of animal husbandry. In this study, we designed and synthesized a novel conjugate florfenicol sulfathiazole (FST) based on drug combination principles, and investigated its antibacterial activity in vitro and its protective effect on inflammatory response and intestinal barrier function in E. coli O78-infected mice in vivo. The results showed that FST had superior antibacterial properties and minimal cytotoxicity compared with its prodrugs as florfenicol and sulfathiazole. FST protected mice from lethal E. coli infection, reduced clinical signs of inflammation, reduced weight loss, alleviated intestinal structural damage. FST decreased the expression of inflammatory cytokines IL-1ß, IL-6, TNF-α, and increased the expression of claudin-1, Occludin, and ZO-1 in the jejunum, improved the intestinal barrier function, and promoted the absorption of nutrients. FST also inhibited the expression of TLR4, MyD88, p-p65, and p-p38 in the jejunum. The study may lay the foundation for the development of FST as new drugs for intestinal inflammation and injury in enteric pathogen infection.


Escherichia coli Infections , Escherichia coli , Thiamphenicol/analogs & derivatives , Humans , Animals , Mice , Intestinal Mucosa , Intestinal Barrier Function , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Escherichia coli Infections/drug therapy , Escherichia coli Infections/pathology , Inflammation/drug therapy , Inflammation/pathology , Sulfathiazole
8.
BMC Vet Res ; 19(1): 262, 2023 Dec 08.
Article En | MEDLINE | ID: mdl-38066606

BACKGROUND: Avian pathogenic Escherichia coli (APEC) causes tracheal damage and heterophilic granulocytic infiltration and inflammation in infected chicks. In this study, we infected chick tracheal tissue with strain AE17 and produced pathological sections with proteomic sequencing. We compared the results of pathological sections from the APEC-infected group with those from the PBS control group; the pathological sections from the experimental group showed hemorrhage, fibrinization, and infiltration of heterophilic granulocytes in the tracheal tissue. In order to explore the effect on proteomics on inflammation and to further search for the caus. RESULTS: The tandem mass tag-based (TMT) sequencing analysis showed 224 upregulated and 140 downregulated proteins after infection with the AE17 strain. Based on the results of KEGG in Complement and coagulation cascades, differential protein expression in the Protein export pathway was upregulated. CONCLUSIONS: With these results, we found that chemokines produced by the Complement and coagulation cascades pathway may cause infiltration of heterophilic granulocytes involved in inflammation, as well as antimicrobial factors produced by the complement system to fight the infection together.These results suggest that APEC causes the infiltration of heterophilic granulocytes through the involvement of the complement system with serine protease inhibitors.


Escherichia coli Infections , Escherichia coli Proteins , Poultry Diseases , Animals , Proteomics , Virulence Factors/metabolism , Escherichia coli Infections/veterinary , Escherichia coli Infections/pathology , Escherichia coli , Chickens/metabolism , Granulocytes , Inflammation/veterinary , Poultry Diseases/pathology
9.
Microb Pathog ; 185: 106435, 2023 Dec.
Article En | MEDLINE | ID: mdl-37931825

Bacterial infections result in intestinal inflammation and injury, which affects gut health and nutrient absorption. Lipocalin 2 (Lcn2) is a protein that reacts to microbial invasion, inflammatory responses, and tissue damage. However, it remains unclear whether Lcn2 has a protective effect against bacterial induced intestinal inflammation. Therefore, this study endeavors to investigate the involvement of Lcn2 in the intestinal inflammation of mice infected with Enterohemorrhagic Escherichia coli O157:H7 (E. coli O157:H7). Lcn2 knockout (Lcn2-/-) mice were used to evaluate the changes of inflammatory responses. Lcn2 deficiency significantly exacerbated clinical symptoms of E. coli O157:H7 infection by reducing body weight and encouraging bacterial colonization of. Compared to infected wild type mice, infected Lcn2-/- mice had significantly elevated levels of pro-inflammatory cytokines in serum and ileum, including interleukin (IL)-6, IL-1ß, and tumor necrosis factor-α (TNF-α), as well as severe villi destruction in the jejunum. Furthermore, Lcn2 deficiency aggravated intestinal barrier degradation by significantly reducing the expression of tight junction proteins occludin and claudin 1, the content of myeloperoxidase (MPO) in the ileum, and the number of goblet cells in the colon. Our findings indicated that Lcn2 could alleviate inflammatory damage caused by E. coli O157:H7 infection in mice by enhancing intestinal barrier function.


Escherichia coli Infections , Escherichia coli O157 , Lipocalin-2 , Animals , Mice , Colon/metabolism , Colon/microbiology , Colon/pathology , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Escherichia coli O157/genetics , Escherichia coli O157/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Lipocalin-2/genetics , Lipocalin-2/metabolism
10.
Microb Pathog ; 184: 106338, 2023 Nov.
Article En | MEDLINE | ID: mdl-37683833

Escherichia coli (E. coli) is a major environmental pathogen that causes mammary tissue damage and cell death, which results in substantial economic losses. Pyroptosis, a novel form of programmed cell death characterized by DNA fragmentation, chromatin condensation, cell swelling and leakage of cell contents, often occurs after inflammatory apoptotic pathways activation. Our objective was to investigate the intraction between E. coli infection and bovine mammary epithelial cells (bMECs) with pyroptosis and to explore the underlying regulatory mechanism. bMECs were infected with E. coli for 6 h. Lactic dehydrogenase activities, interleukin (IL)-10, IL-1ß, IL-18 and tumor necrosis factor-α concentrations, total apoptosis indexes, and protein expressions of P-cdc25c, P-CDK1, cleaved caspase 9, cleaved caspase 3, cleaved PARP, P-NF-κB, NLRP3, ASC, caspase 1, gasdermin D N-terminal, IL-1ß and IL-18 were significantly increased in E. coli infected bMECs. Whereas, cell membrane potential, protein levels of cdc25c, CDK1, cyclin B1, and Bcl-2/Bax level were markedly reduced. Furthermore, Ac-DEVD-CHO (specific inhibitor of apoptosis) dramatically suppressed pyroptosis in bMECs. Moreover, expressions of p53 and p21 promptly improved after E. coli infection, however, Pifithrin-α (specific inhibitor of p53) inhibited p53-p21 pathway, apoptosis, cell cycle arrest and pyroptosis. These results elaborated that E. coli infection of bMECs induced pyroptosis through activating the p53-p21 pathway-mediated apoptosis and cell cycle arrest. Taken together, inhibition of pyroptosis via suppressing of p53-p21 pathway may be an effective therapeutic approach for treating E. coli-induced mastitis, offering efficient theoretical support for the protection and treatment of bovine mastitis.


Escherichia coli Infections , Pyroptosis , Female , Cattle , Animals , Interleukin-18/metabolism , Tumor Suppressor Protein p53/metabolism , Escherichia coli/metabolism , Apoptosis , Epithelial Cells , Escherichia coli Infections/pathology , Cell Cycle Checkpoints
11.
J Microbiol ; 61(8): 715-727, 2023 Aug.
Article En | MEDLINE | ID: mdl-37665555

Enterohemorrhagic Escherichia coli (EHEC) is a specific subset of Shiga toxin-producing Escherichia coli (STEC) strains that are characterized by their ability to cause bloody diarrhea (hemorrhagic colitis) and potentially life-threatening, extraintestinal complications such as hemolytic uremic syndrome (HUS), which is associated with acute renal failure., contributing to severe clinical outcomes. The Shiga toxins (Stxs), produced by EHEC, are primary virulence factors. These potent cytotoxins are composed of one enzymatically active A subunit (StxA) and five receptor-binding B subunits (StxB). Although the toxins are primarily associated with cytotoxic effects, they also elicit other pathogenic consequences due to their induction of a number of biological processes, including apoptosis through ER-stress, pro-inflammatory responses, autophagy, and post-translational modification (PTM). Moreover, several studies have reported the association between Stxs and extracellular vesicles (EVs), including microvesicles and exosomes, demonstrating that Stx-containing EVs secreted by intoxicated macrophages are taken up by recipient cells, such as toxin-sensitive renal proximal tubular epithelial cells. This mechanism likely contributes to the spreading of Stxs within the host, and may exacerbate gastrointestinal illnesses and kidney dysfunction. In this review, we summarize recent findings relating to the host responses, in different types of cells in vitro and in animal models, mediated by Stxs-containing exosomes. Due to their unique properties, EVs have been explored as therapeutic agents, drug delivery systems, and diagnostic tools. Thus, potential therapeutic applications of EVs in EHEC Stxs-mediated pathogenesis are also briefly reviewed.


Enterohemorrhagic Escherichia coli , Escherichia coli Infections , Extracellular Vesicles , Hemolytic-Uremic Syndrome , Shiga-Toxigenic Escherichia coli , Animals , Shiga Toxin , Shiga Toxins/toxicity , Escherichia coli Infections/pathology
12.
J Crohns Colitis ; 17(12): 1988-2001, 2023 Dec 30.
Article En | MEDLINE | ID: mdl-37462681

IFNγ-producing ex-Th17 cells ['Th1/17'] were shown to play a key pathogenic role in experimental colitis and are abundant in the intestine. Here, we identified and characterised a novel, potentially colitogenic subset of Th17 cells in the intestine of patients with Crohn's disease [CD]. Human Th17 cells expressing CCR5 ['pTh17'] co-expressed T-bet and RORC/γt and produced very high levels of IL-17, together with IFN-γ. They had a gene signature of Th17 effector cells and were distinct from established Th1/17 cells. pTh17 cells, but not Th1/17 cells, were associated with intestinal inflammation in CD, and decreased upon successful anti-TNF therapy with infliximab. Conventional CCR5[-]Th17 cells differentiated to pTh17 cells with IL-23 in vitro. Moreover, anti-IL-23 therapy with risankizumab strongly reduced pTh17 cells in the intestine. Importantly, intestinal pTh17 cells were selectively activated by adherent-invasive Escherichia coli [AIEC], but not by a commensal/probiotic E. coli strain. AIEC induced high levels of IL-23 and RANTES from dendritic cells [DC]. Intestinal CCR5+Th1/17 cells responded instead to cytomegalovirus and were reduced in ulcerative colitis [UC], suggesting an unexpected protective role. In conclusion, we identified an IL-23-inducible subset of human intestinal Th17 cells. pTh17 cells produced high levels of pro-inflammatory cytokines, were selectively associated with intestinal inflammation in CD, and responded to CD-associated AIEC, suggesting a key colitogenic role.


Crohn Disease , Escherichia coli Infections , Humans , Crohn Disease/pathology , Escherichia coli , Th17 Cells/pathology , Tumor Necrosis Factor Inhibitors , Intestines/pathology , Inflammation/pathology , Escherichia coli Infections/complications , Escherichia coli Infections/pathology , Interleukin-23 , Intestinal Mucosa/pathology , Bacterial Adhesion
13.
Nat Commun ; 14(1): 3667, 2023 06 20.
Article En | MEDLINE | ID: mdl-37339949

The intrinsic virulence of extra-intestinal pathogenic Escherichia coli is associated with numerous chromosomal and/or plasmid-borne genes, encoding diverse functions such as adhesins, toxins, and iron capture systems. However, the respective contribution to virulence of those genes seems to depend on the genetic background and is poorly understood. Here, we analyze genomes of 232 strains of sequence type complex STc58 and show that virulence (quantified in a mouse model of sepsis) emerged in a sub-group of STc58 due to the presence of the siderophore-encoding high-pathogenicity island (HPI). When extending our genome-wide association study to 370 Escherichia strains, we show that full virulence is associated with the presence of the aer or sit operons, in addition to the HPI. The prevalence of these operons, their co-occurrence and their genomic location depend on strain phylogeny. Thus, selection of lineage-dependent specific associations of virulence-associated genes argues for strong epistatic interactions shaping the emergence of virulence in E. coli.


Escherichia coli Infections , Escherichia coli , Animals , Mice , Virulence/genetics , Iron , Escherichia coli Infections/pathology , Genomic Islands/genetics , Genome-Wide Association Study , Phylogeny
14.
Anim Biotechnol ; 34(8): 3681-3692, 2023 Dec.
Article En | MEDLINE | ID: mdl-37083115

Colibacillosis is a complex disease that caused by avian pathogenic Escherichia coli (APEC), resulting in huge economic loss to the global poultry industry and threatening to human health. Alternative splicing (AS) is a universal post-transcriptional regulatory mechanism, which can simultaneously produce many proteins from a single gene to involve in various diseases and individual development. Herein, we characterized genome-wide AS events in wild type macrophages (WT) and APEC infected macrophages (APEC) by high-throughput RNA sequencing technology. A total of 751 differentially expressed (DE) AS genes were identified in the comparison of APEC vs. WT, including 587 of SE, 114 of MXE, 25 of RI, 17 of A3 and 8 of A5 event. Functional analysis showed that these identified DE AS genes were involved in 'Endocytosis', 'p53 signaling pathway', 'MAPK signaling pathway', 'NOD-like receptor signaling pathway', 'Ubiquitin mediated proteolysis' and 'Focal adhesion' immune related pathways. In summary, we comprehensively investigate AS events during APEC infection. This study has expanded our understanding of the process of APEC infection and provided new insights for further treatment options for APEC infection.


Escherichia coli Infections , Poultry Diseases , Animals , Humans , Escherichia coli/genetics , Chickens/genetics , Alternative Splicing/genetics , Escherichia coli Infections/genetics , Escherichia coli Infections/veterinary , Escherichia coli Infections/pathology , Macrophages/metabolism , Macrophages/pathology , Poultry Diseases/genetics , Poultry Diseases/metabolism , Poultry Diseases/pathology
15.
Vet Pathol ; 60(3): 336-340, 2023 05.
Article En | MEDLINE | ID: mdl-36951102

This case report describes a case of granulomatous colitis (GC) associated with adherent-invasive Escherichia coli (AIEC) with extension to cecum and ileum and dissemination to multiple lymph nodes, the spleen, and brain in a 10-year-old, male Sphynx cat. The cat had an episode of diarrhea 4 months prior to consultation due to sudden blindness. Signs rapidly progressed to ataxia, seizures, and death. Gross and histologic findings were consistent with granulomatous inflammation in all affected organs. In situ hybridization confirmed the presence of intracellular E. coli within enterocytes and infiltrating macrophages, and whole genome sequencing identified virulence traits commonly linked to AIEC strain. This is the first characterization of GC in a cat associated to AIEC resembling the metastatic form of Crohn's disease in humans and GC of dogs. Extraintestinal involvement might provide evidence of the ability of AIEC to promote granulomatous inflammation beyond the gut.


Crohn Disease , Dog Diseases , Escherichia coli Infections , Humans , Male , Animals , Dogs , Crohn Disease/complications , Crohn Disease/pathology , Crohn Disease/veterinary , Escherichia coli/genetics , Escherichia coli Infections/etiology , Escherichia coli Infections/pathology , Escherichia coli Infections/veterinary , Intestinal Mucosa/pathology , Inflammation/pathology , Inflammation/veterinary , Bacterial Adhesion/genetics , Dog Diseases/pathology
16.
Gut Microbes ; 15(1): 2193115, 2023.
Article En | MEDLINE | ID: mdl-36945126

The interaction between adherent-invasive Escherichia coli (AIEC) and intestinal macrophages is implicated in the pathogenesis of Crohn's disease (CD). However, its role in intestinal fibrogenesis and the underlying molecular mechanisms are poorly understood. In addition, miRNAs such as let-7b may participate in AIEC-macrophage interactions. In this study, we identified that the colonization of AIEC in the ileum was associated with enhanced intestinal fibrosis and reduced let-7b expression by enrolling a prospective cohort of CD patients undergoing ileocolectomy. Besides, AIEC-infected IL-10-/- mice presented more severe intestinal fibrosis and could be improved by exogenous let-7b. Mechanistically, intestinal macrophages were found to be the main target of let-7b. Transferring let-7b-overexpressing macrophages to AIEC-infected IL-10-/- mice significantly alleviated intestinal fibrosis. In vitro, AIEC suppressed exosomal let-7b derived from intestinal epithelial cells (IECs), instead of the direct inhibition of let-7b in macrophages, to promote macrophages to a fibrotic phenotype. Finally, TGFßR1 was identified as one target of let-7b that regulates macrophage polarization. Overall, the results of our work indicate that AIEC is associated with enhanced intestinal fibrosis in CD. AIEC could inhibit exosomal let-7b from IECs to promote intestinal macrophages to a fibrotic phenotype and then contributed to fibrogenesis. Thus, anti-AIEC or let-7b therapy may serve as novel therapeutic approaches to ameliorate intestinal fibrosis.


What is the context?Adherent-invasive Escherichia coli (AIEC) plays an important role in the pathogenesis of Crohn's disease (CD) and has a strong association with intestinal fibrosis in animal models. However, how these bacteria contribute to intestinal fibrosis in CD patients is still unclear.The plasticity of macrophages is crucial in immune tolerance and tissue repair in the gastrointestinal tract, and the abnormal interaction between macrophages and gut bacteria triggers the fibrogenesis in the intestine.The association between the miRNA let-7b and fibrosis process has been widely reported in many tissues, except the intestine.What is new?AIEC colonization in the terminal ileum is associated with severe intestinal fibrosis in CD patients and the let-7b plays an anti-fibrotic role in intestinal fibrosis.Intestinal macrophages are the key modulator of AIEC-induced fibrosis and can be promoted to an antifibrotic phenotype through let-7b-targeted TGFßR1 inhibition.AIEC suppresses intestinal epithelial cell-derived exosomal let-7b to promote intestinal macrophages to a fibrotic phenotype, rather than a direct effect on macrophage regulation.What is the impact? Anti-AIEC and let-7b therapy may serve as potential therapeutic strategies to reduce intestinal fibrosis in CD in the future.


Crohn Disease , Escherichia coli Infections , Gastrointestinal Microbiome , Animals , Mice , Bacterial Adhesion , Crohn Disease/pathology , Epithelial Cells/metabolism , Escherichia coli/metabolism , Escherichia coli Infections/pathology , Fibrosis , Interleukin-10/genetics , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Macrophages/metabolism , Prospective Studies , Humans
17.
Virulence ; 14(1): 2150453, 2023 12.
Article En | MEDLINE | ID: mdl-36411420

Avian pathogenic Escherichia coli (APEC) leads to economic losses in poultry industry and is also a threat to human health. Various strategies were used for searching virulence factors, while little is known about the mechanism by which APEC survives in host or is eliminated by host. Thus, chicken colibacillosis model was constructed by intraperitoneally injecting E. coli O78 in this study, then the protein dynamic expression of spleen was characterized at different post-infection times by quantitative proteome. Comparative analysis showed that E. coli induced significant dysregulation at 72 h post infection in spleen tissue. Transcriptomic method was further used to assess the changes of dysregulated proteins at 72 h post infection at the mRNA level. Total 278 protein groups (5.7%) and 2,443 genes (24.4%) were dysregulated, respectively. The upregulated proteins and genes were consistently enriched in phagosome and lysosome pathways, indicating E. coli infection activates phagosome maturation pathway. The matured phagolysosome might kill the invasive E. coli. This study illuminated the genetic dysregulation in chicken spleen at the protein and mRNA levels after E. coli infecting and identified candidate genes for host response to APEC infection.


Escherichia coli Infections , Poultry Diseases , Proteogenomics , Animals , Chickens , Escherichia coli/metabolism , Escherichia coli Infections/veterinary , Escherichia coli Infections/pathology , Phagosomes , Poultry Diseases/microbiology , Poultry Diseases/pathology , Spleen/pathology
18.
Avian Pathol ; 52(1): 62-77, 2023 Feb.
Article En | MEDLINE | ID: mdl-36399118

Avian pathogenic E. coli (APEC) can cause localized and systemic diseases in poultry, threatening human health via meat or egg contamination and resulting in considerable economic losses to the poultry industry globally. Increasing evidence shows circRNAs were widely involved in various biological processes. However, the role of circRNAs in the host response against APEC infection, especially correlated with the regulation of RIP2, remains unclear. Herein, the RNAseq technology was used to identify the circRNA expression profiles in the overexpression of RIP2 macrophages with or without APEC infection. A total of 256 and 287 differentially expressed (DE) circRNAs were identified in the overexpression of RIP2 group (oeRIP2) vs. the wild-type group (WT) and oeRIP2 + APEC vs. APEC, respectively, whose parental genes were involved in MAPK signalling pathway, Wnt signalling pathway, focal adhesion, tight junction, and VEGF signalling pathways. Specifically, the key circRNAs, such as 5:814443-825127, 10:18922360-18928461, 2:8746306-8750639, and 2:124177751-124184063 might play a critical role in APEC infection and the regulation of RIP2. As a whole, these findings will facilitate understanding the molecular mechanism underlying circRNAs, especially related to the regulation of the RIP2 gene. Meanwhile, the study may offer new ideas to improve host immune and inflammatory response against APEC infection.


Escherichia coli Infections , Poultry Diseases , Animals , Humans , Chickens/genetics , Escherichia coli/genetics , RNA, Circular/metabolism , Escherichia coli Infections/veterinary , Escherichia coli Infections/pathology , Macrophages , Poultry Diseases/pathology
19.
Cell Mol Neurobiol ; 43(5): 2203-2217, 2023 Jul.
Article En | MEDLINE | ID: mdl-36227397

Shiga toxin 2 (Stx2) from enterohemorrhagic Escherichia coli (EHEC) produces hemorrhagic colitis, hemolytic uremic syndrome (HUS), and acute encephalopathy. The mortality rate in HUS increases significantly when the central nervous system (CNS) is involved. Besides, EHEC also releases lipopolysaccharide (LPS). Many reports have described cognitive dysfunctions in HUS patients, the hippocampus being one of the brain areas targeted by EHEC infection. In this context, a translational murine model of encephalopathy was employed to establish the deleterious effects of Stx2 and the contribution of LPS in the hippocampus. The purpose of this work is to elucidate the signaling pathways that may activate the inflammatory processes triggered by Stx2, which produces cognitive alterations at the level of the hippocampus. Results demonstrate that Stx2 produced depression-like behavior, pro-inflammatory cytokine release, and NF-kB activation independent of the ERK1/2 signaling pathway, while co-administration of Stx2 and LPS reduced memory index. On the other hand, LPS activated NF-kB dependent on ERK1/2 signaling pathway. Cotreatment of Stx2 with LPS aggravated the pathologic state, while dexamethasone treatment succeeded in preventing behavioral alterations. Our present work suggests that the use of drugs such as corticosteroids or NF-kB signaling inhibitors may serve as neuroprotectors from EHEC infection.


Brain Diseases , Cognitive Dysfunction , Enterohemorrhagic Escherichia coli , Escherichia coli Infections , Hemolytic-Uremic Syndrome , Mice , Humans , Animals , Shiga Toxin 2/toxicity , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System , NF-kappa B , Brain/pathology , Escherichia coli Infections/complications , Escherichia coli Infections/drug therapy , Escherichia coli Infections/pathology , Hippocampus/pathology , Cognition
20.
Anim Biotechnol ; 34(7): 3267-3273, 2023 Dec.
Article En | MEDLINE | ID: mdl-36007588

To understand the pathology of natural cases of E. coli pathotypes infection in bovine calves, 45 cases of bovine calves, below one month of age, died due to enteritis were studied. Total seventeen cases (37.77%) turned positive for different pathotypes of E. coli by RT-PCR. Out of seventeen positive samples for E. coli, six cases (35.29%) were positive for eae gene, three cases (17.64%) for bfp gene and eight cases (47.05) for fimA gene of E. coli. Gross lesions in these cases showed pin-point to ecchymotic hemorrhages in the mucosa of jejunum, ileum and colon. The draining mesenteric lymph nodes were swollen, enlarged and showed cord -like structure. Histopathology of small intestine showed, villi lining cells were sloughed off, tips of villi capillary plexus were congested and hemorrhagic, and skipping lesions of microabscesses in the crypts of mucosa were observed. In the duodenum, necrosis of crypts and infiltration of mononuclear cells in the lamina propria and around Brunner's gland. In mesenteric lymph nodes the subscapular space were infiltrated with mononuclear cells with depletion of lymphoid follicles in cortical area. Peri-trabecular and medullary sinuses of mesenteric lymph nodes were necrosed.


Cattle Diseases , Escherichia coli Infections , Animals , Cattle , Escherichia coli/genetics , Diarrhea/veterinary , Diarrhea/pathology , Escherichia coli Infections/veterinary , Escherichia coli Infections/pathology , Ileum/pathology , Jejunum/pathology
...