Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 31
1.
Mar Drugs ; 20(3)2022 Mar 17.
Article En | MEDLINE | ID: mdl-35323512

Four new dimeric sorbicillinoids (1-3 and 5) and a new monomeric sorbicillinoid (4) as well as six known analogs (6-11) were purified from the fungal strain Hypocrea jecorina H8, which was obtained from mangrove sediment, and showed potent inhibitory activity against the tea pathogenic fungus Pestalotiopsis theae (P. theae). The planar structures of 1-5 were assigned by analyses of their UV, IR, HR-ESI-MS, and NMR spectroscopic data. All the compounds were evaluated for growth inhibition of tea pathogenic fungus P. theae. Compounds 5, 6, 8, 9, and 10 exhibited more potent inhibitory activities compared with the positive control hexaconazole with an ED50 of 24.25 ± 1.57 µg/mL. The ED50 values of compounds 5, 6, 8, 9, and 10 were 9.13 ± 1.25, 2.04 ± 1.24, 18.22 ± 1.29, 1.83 ± 1.37, and 4.68 ± 1.44 µg/mL, respectively. Additionally, the effects of these compounds on zebrafish embryo development were also evaluated. Except for compounds 5 and 8, which imparted toxic effects on zebrafish even at 0.625 µM, the other isolated compounds did not exhibit significant toxicity to zebrafish eggs, embryos, or larvae. Taken together, sorbicillinoid derivatives (6, 9, and 10) from H. jecorina H8 displayed low toxicity and high anti-tea pathogenic fungus potential.


Ascomycota/drug effects , Biological Control Agents , Hypocreales/chemistry , Polyketides , Animals , Ascomycota/growth & development , Biological Control Agents/chemistry , Biological Control Agents/isolation & purification , Biological Control Agents/pharmacology , Biological Control Agents/toxicity , Camellia sinensis/microbiology , Embryo, Nonmammalian , Molecular Structure , Polyketides/chemistry , Polyketides/isolation & purification , Polyketides/pharmacology , Polyketides/toxicity , Zebrafish
2.
Toxins (Basel) ; 13(12)2021 12 14.
Article En | MEDLINE | ID: mdl-34941734

Colibactin is a genotoxin produced primarily by Escherichia coli harboring the genomic pks island (pks+ E. coli). Pks+ E. coli cause host cell DNA damage, leading to chromosomal instability and gene mutations. The signature of colibactin-induced mutations has been described and found in human colorectal cancer (CRC) genomes. An inflamed intestinal environment drives the expansion of pks+ E. coli and promotes tumorigenesis. Mesalamine (i.e., 5-aminosalycilic acid), an effective anti-inflammatory drug, is an inhibitor of the bacterial polyphosphate kinase (PPK). This drug not only inhibits the production of intestinal inflammatory mediators and the proliferation of CRC cells, but also limits the abundance of E. coli in the gut microbiota and diminishes the production of colibactin. Here, we describe the link between intestinal inflammation and colorectal cancer induced by pks+ E. coli. We discuss the potential mechanisms of the pleiotropic role of mesalamine in treating both inflammatory bowel diseases and reducing the risk of CRC due to pks+ E. coli.


Escherichia coli/metabolism , Mesalamine/pharmacology , Neoplasms/chemically induced , Neoplasms/prevention & control , Peptides/toxicity , Polyketides/toxicity , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Humans
3.
Cancer Cell ; 39(11): 1439-1441, 2021 11 08.
Article En | MEDLINE | ID: mdl-34752751

Gut colonization by colibactin-producing bacteria is associated with colorectal cancer. A mutational signature of this genotoxin in human cancer indicates causality but only partially accounts for cell transformation. Instead, the failure of adequately resolving DNA damage causes genomic aberrations and chromosomal instability, constituting the main starting point for colibactin-driven cancer.


Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/genetics , Peptides/toxicity , Polyketides/toxicity , Cell Transformation, Neoplastic/chemically induced , Chromosome Aberrations , Colorectal Neoplasms/chemically induced , DNA Damage , Humans , Mutation
4.
Toxins (Basel) ; 13(8)2021 08 06.
Article En | MEDLINE | ID: mdl-34437417

Colibactin is a complex secondary metabolite that leads to genotoxicity that interferes with the eukaryotic cell cycle. It plays an important role in many diseases, including neonatal mouse sepsis and meningitis. Avian pathogenic Escherichia coli (APEC) is responsible for several diseases in the poultry industry and may threaten human health due to its potential zoonosis. In this study, we confirmed that clbG was necessary for the APEC XM strain to produce colibactin. The deletion of clbG on APEC XM contributed to lowered γH2AX expression, no megalocytosis, and no cell cycle arrest in vitro. None of the 4-week Institute of Cancer Research mice infected with the APEC XM ΔclbG contracted meningitis or displayed weakened clinical symptoms. Fewer histopathological lesions were observed in the APEC XM ΔclbG group. The bacterial colonization of tissues and the relative expression of cytokines (IL-1ß, IL-6, and TNF-α) in the brains decreased significantly in the APEC XM ΔclbG group compared to those in the APEC XM group. The tight junction proteins (claudin-5, occludin, and ZO-1) were not significantly destroyed in APEC XM ΔclbG group in vivo and in vitro. In conclusion, clbG is necessary for the synthesis of the genotoxin colibactin and affects the development of APEC meningitis in mice.


Escherichia coli Infections , Peptides/toxicity , Polyketides/toxicity , Animals , Brain/metabolism , Brain/pathology , Cell Line , Cytokines/genetics , Disease Models, Animal , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Escherichia coli Infections/veterinary , Female , Male , Mice, Inbred ICR , Peptides/genetics , Peptides/metabolism , Polyketides/metabolism , Poultry Diseases , Tight Junction Proteins/metabolism
5.
Toxins (Basel) ; 13(5)2021 05 12.
Article En | MEDLINE | ID: mdl-34065799

Colibactin is a secondary metabolite encoded by the pks gene island identified in several Enterobacteriaceae, including some pathogenic Escherichia coli (E. coli) commonly enriched in mucosal tissue collected from patients with inflammatory bowel disease and colorectal cancer. E. coli harboring this biosynthetic gene cluster cause DNA damage and tumorigenesis in cell lines and pre-clinical models, yet fundamental knowledge regarding colibactin function is lacking. To accurately assess the role of pks+ E. coli in cancer etiology, the biological mechanisms governing production and delivery of colibactin by these bacteria must be elucidated. In this review, we will focus on recent advances in our understanding of colibactin's structural mode-of-action and mutagenic potential with consideration for how this activity may be regulated by physiologic conditions within the intestine.


Enterobacteriaceae/metabolism , Mutagens/metabolism , Peptides/metabolism , Polyketides/metabolism , Animals , DNA Damage , Enterobacteriaceae/genetics , Humans , Multigene Family , Mutagens/toxicity , Peptides/toxicity , Polyketides/toxicity , Secondary Metabolism
6.
Toxicon ; 197: 126-135, 2021 Jul 15.
Article En | MEDLINE | ID: mdl-33901549

Microbiota can significantly contribute to colorectal cancer initiation and development. It was described that E. coli harbouring polyketide synthase (pks) genes can synthetize bacterial toxin colibactin, which was first described by Nougayrede's group in 2006. E. coli positive for pks genes were overrepresented in colorectal cancer biopsies and, therefore, prevalence and the effect of pks positive bacteria as a risk factor in colorectal cancer development is in our interest. Interestingly, pks gene cluster in E. coli shares a striking 100% sequence identity with K. pneumoniae, suggesting that their function and regulation are conserved. Moreover, K. pneumoniae can express a variety of virulence factors, including capsules, siderophores, iron-scavenging systems, adhesins and endotoxins. It was reported that pks cluster and thereby colibactin is also related to the hypervirulence of K. pneumoniae. Acquisition of the pks locus is associated with K. pneumoniae gut colonisation and mucosal invasion. Colibactin also increases the likelihood of serious complications of bacterial infections, such as development of meningitis and potentially tumorigenesis. Even though K. pneumoniae is undoubtedly a gut colonizer, the role of pks positive K. pneumoniae in GIT has not yet been investigated. It seems that CRC-distinctive microbiota is already present in the early stages of cancer development and, therefore, microbiome analysis could help to discover the early stages of cancer, which are crucial for effectiveness of anticancer therapy. We hypothesize, that pks positive K. pneumoniae can be a potential biomarker of tumour prevalence and anticancer therapy response.


Bacterial Toxins , Colorectal Neoplasms , Polyketides , Colorectal Neoplasms/chemically induced , Escherichia coli , Humans , Klebsiella pneumoniae , Peptides , Polyketides/toxicity
7.
Chirality ; 32(3): 299-307, 2020 03.
Article En | MEDLINE | ID: mdl-31975445

(±)-Pratenone A (1), the first representative of natural 3-(1-naphthyl)-2-benzofuran-1(3H)-one polyketides, was isolated from a marine-derived Streptomyces pratensis strain KCB-132 together with three other new analogues (2-4). Its structure was assigned by spectroscopic analysis, and the absolute configurations of the two enantiomers separated by high-performance liquid chromatography were determined by single-crystal X-ray diffraction and electronic circular dichroism calculations. The solvent-induced racemization of 1 and a proposed biogenetic pathway to 1-4 from the co-isolated angucyclinone precursor, as well as their biological activity, are also discussed.


Polyketides/chemistry , Streptomyces/chemistry , Anthraquinones/chemistry , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Aquatic Organisms , Benzofurans/chemistry , Chromatography, High Pressure Liquid , Circular Dichroism , Crystallography, X-Ray , HL-60 Cells , Hep G2 Cells , Humans , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Molecular Structure , Polyketides/isolation & purification , Polyketides/pharmacology , Polyketides/toxicity , Staphylococcus aureus/drug effects , Stereoisomerism , Streptomyces/isolation & purification
8.
Gastroenterology ; 158(5): 1373-1388, 2020 04.
Article En | MEDLINE | ID: mdl-31917256

BACKGROUND & AIMS: Colibactin-producing Escherichia coli (CoPEC) colonize the colonic mucosa of a higher proportion of patients with vs without colorectal cancer (CRC) and promote colorectal carcinogenesis in susceptible mouse models of CRC. Autophagy degrades cytoplasmic contents, including intracellular pathogens, via lysosomes and regulates intestinal homeostasis. We investigated whether inhibiting autophagy affects colorectal carcinogenesis in susceptible mice infected with CoPEC. METHODS: Human intestinal epithelial cells (IECs) (HCT-116) were infected with a strain of CoPEC (11G5 strain) isolated from a patient or a mutant strain that does not produce colibactin (11G5ΔclbQ). Levels of ATG5, ATG16L1, and SQSTM1 (also called p62) were knocked down in HCT-116 cells using small interfering RNAs. ApcMin/+ mice and ApcMin/+ mice with IEC-specific disruption of Atg16l1 (ApcMin/+/Atg16l1ΔIEC) were infected with 11G5 or 11G5ΔclbQ. Colonic tissues were collected from mice and analyzed for tumor size and number and by immunohistochemical staining, immunoblot, and quantitative reverse transcription polymerase chain reaction for markers of autophagy, DNA damage, cell proliferation, and inflammation. We analyzed levels of messenger RNAs (mRNAs) encoding proteins involved in autophagy in colonic mucosal tissues from patients with sporadic CRC colonized with vs without CoPEC by quantitative reverse-transcription polymerase chain reaction. RESULTS: Patient colonic mucosa with CoPEC colonization had higher levels of mRNAs encoding proteins involved in autophagy than colonic mucosa without these bacteria. Infection of cultured IECs with 11G5 induced autophagy and DNA damage repair, whereas infection with 11G5ΔclbQ did not. Knockdown of ATG5 in HCT-116 cells increased numbers of intracellular 11G5, secretion of interleukin (IL) 6 and IL8, and markers of DNA double-strand breaks but reduced markers of DNA repair, indicating that autophagy is required for bacteria-induced DNA damage repair. Knockdown of ATG5 in HCT-116 cells increased 11G5-induced senescence, promoting proliferation of uninfected cells. Under uninfected condition, ApcMin/+/Atg16l1ΔIEC mice developed fewer and smaller colon tumors than ApcMin/+ mice. However, after infection with 11G5, ApcMin/+/Atg16l1ΔIEC mice developed more and larger tumors, with a significant increase in mean histologic score, than infected ApcMin/+ mice. Increased levels of Il6, Tnf, and Cxcl1 mRNAs, decreased level of Il10 mRNA, and increased markers of DNA double-strand breaks and proliferation were observed in the colonic mucosa of 11G5-infected ApcMin/+/Atg16l1ΔIEC mice vs 11G5-infected ApcMin/+ mice. CONCLUSION: Infection of IECs and susceptible mice with CoPEC promotes autophagy, which is required to prevent colorectal tumorigenesis. Loss of ATG16L1 from IECs increases markers of inflammation, DNA damage, and cell proliferation and increases colorectal tumorigenesis in 11G5-infected ApcMin/+ mice. These findings indicate the importance of autophagy in response to CoPEC infection, and strategies to induce autophagy might be developed for patients with CRC and CoPEC colonization.


Autophagy , Carcinogenesis/immunology , Colon/microbiology , Colonic Neoplasms/immunology , Intestinal Mucosa/microbiology , Adenomatous Polyposis Coli Protein/genetics , Animals , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/immunology , Autophagy-Related Proteins/metabolism , Carcinogenesis/drug effects , Cell Proliferation , Colon/immunology , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/microbiology , Colonic Neoplasms/pathology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Escherichia coli/immunology , Escherichia coli/isolation & purification , Escherichia coli/pathogenicity , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HCT116 Cells , HeLa Cells , Host-Pathogen Interactions/immunology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Mice, Transgenic , Peptides/toxicity , Polyketides/toxicity , RNA, Small Interfering/metabolism
9.
J Toxicol Sci ; 44(12): 871-876, 2019.
Article En | MEDLINE | ID: mdl-31813906

Colibactin is a polyketide-peptide genotoxin produced by enteric bacteria such as E. coli, and is considered to contribute to the development of colorectal cancer. We previously isolated E. coli strains from Japanese colorectal cancer patients, and in the present study we investigated the genotoxic potency of the colibactin-producing (clb+) E. coli strains that carry the polyketide synthases "pks" gene cluster (pks+) and an isogenic clb- mutant in which the colibactin-producing ability is impaired. Measurement of phosphorylated histone H2AX indicated that DNA double strand breaks were induced in mammalian CHO AA8 cells infected with the clb+ E. coli strains. Induction of DNA damage response (SOS response) by crude extract of the clb+ strains was 1.7 times higher than that of the clb- E. coli in an umu assay with a Salmonella typhimurium TA1535/pSK1002 tester strain. Micronucleus test with CHO AA8 cells revealed that infection with the clb+ strains induced genotoxicity, i.e., the frequencies of micronucleated cells infected with clb+ strain were 4-6 times higher than with the clb- strain. Since the intestinal flora are affected by dietary habits that are strongly associated with ethnicity, these data may contribute to both risk evaluation and prevention of colorectal cancer in the Japanese population.


Colon/microbiology , Colorectal Neoplasms/microbiology , Escherichia coli/isolation & purification , Mutagens/toxicity , Peptides/toxicity , Polyketides/toxicity , Aged , Animals , CHO Cells , Cricetulus , DNA Breaks, Double-Stranded/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Male , Micronuclei, Chromosome-Defective/chemically induced , Mutagens/metabolism , Peptides/metabolism , Polyketides/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics
10.
PLoS Pathog ; 15(9): e1007921, 2019 09.
Article En | MEDLINE | ID: mdl-31568537

Humans are frequently exposed to bacterial genotoxins involved in digestive cancers, colibactin and Cytolethal Distending Toxin (CDT), the latter being secreted by many pathogenic bacteria. Our aim was to evaluate the effects induced by these genotoxins on nuclear remodeling in the context of cell survival. Helicobacter infected mice, coculture experiments with CDT- and colibactin-secreting bacteria and hepatic, intestinal and gastric cells, and xenograft mouse-derived models were used to assess the nuclear remodeling in vitro and in vivo. Our results showed that CDT and colibactin induced-nuclear remodeling can be associated with the formation of deep cytoplasmic invaginations in the nucleus of giant cells. These structures, observed both in vivo and in vitro, correspond to nucleoplasmic reticulum (NR). The core of the NR was found to concentrate ribosomes, proteins involved in mRNA translation, polyadenylated RNA and the main components of the complex mCRD involved in mRNA turnover. These structures are active sites of mRNA translation, correlated with a high degree of ploidy, and involve MAPK and calcium signaling. Additional data showed that insulation and concentration of these adaptive ribonucleoprotein particles within the nucleus are dynamic, transient and protect the cell until the genotoxic stress is relieved. Bacterial genotoxins-induced NR would be a privileged gateway for selected mRNA to be preferably transported therein for local translation. These findings offer new insights into the context of NR formation, a common feature of many cancers, which not only appears in response to therapies-induced DNA damage but also earlier in response to genotoxic bacteria.


Bacterial Toxins/toxicity , Helicobacter/pathogenicity , Ribonucleoproteins/metabolism , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Survival , DNA Damage , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mutagens/toxicity , Peptides/toxicity , Polyketides/toxicity , RNA, Messenger/metabolism
11.
PLoS Pathog ; 15(9): e1008029, 2019 09.
Article En | MEDLINE | ID: mdl-31545853

Although Escherichia coli Nissle 1917 (EcN) has been used therapeutically for over a century, the determinants of its probiotic properties remain elusive. EcN produces two siderophore-microcins (Mcc) responsible for an antagonistic activity against other Enterobacteriaceae. EcN also synthesizes the genotoxin colibactin encoded by the pks island. Colibactin is a virulence factor and a putative pro-carcinogenic compound. Therefore, we aimed to decouple the antagonistic activity of EcN from its genotoxic activity. We demonstrated that the pks-encoded ClbP, the peptidase that activates colibactin, is required for the antagonistic activity of EcN. The analysis of a series of ClbP mutants revealed that this activity is linked to the transmembrane helices of ClbP and not the periplasmic peptidase domain, indicating the transmembrane domain is involved in some aspect of Mcc biosynthesis or secretion. A single amino acid substitution in ClbP inactivates the genotoxic activity but maintains the antagonistic activity. In an in vivo salmonellosis model, this point mutant reduced the clinical signs and the fecal shedding of Salmonella similarly to the wild type strain, whereas the clbP deletion mutant could neither protect nor outcompete the pathogen. The ClbP-dependent antibacterial effect was also observed in vitro with other E. coli strains that carry both a truncated form of the Mcc gene cluster and the pks island. In such strains, siderophore-Mcc synthesis also required the glucosyltransferase IroB involved in salmochelin production. This interplay between colibactin, salmochelin, and siderophore-Mcc biosynthetic pathways suggests that these genomic islands were co-selected and played a role in the evolution of E. coli from phylogroup B2. This co-evolution observed in EcN illustrates the fine margin between pathogenicity and probiotic activity, and the need to address both the effectiveness and safety of probiotics. Decoupling the antagonistic from the genotoxic activity by specifically inactivating ClbP peptidase domain opens the way to the safe use of EcN.


Escherichia coli/physiology , Mutagens/toxicity , Probiotics/therapeutic use , Animals , Antibiosis/genetics , Antibiosis/physiology , Bacteriocins/genetics , Bacteriocins/metabolism , Bacteriocins/toxicity , Biosynthetic Pathways/genetics , Enterobactin/analogs & derivatives , Enterobactin/genetics , Enterobactin/physiology , Enterobactin/toxicity , Escherichia coli/genetics , Escherichia coli/pathogenicity , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/physiology , Female , Genes, Bacterial , Genomic Islands , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Multigene Family , Mutation , Peptide Hydrolases/chemistry , Peptide Hydrolases/genetics , Peptide Hydrolases/physiology , Peptides/genetics , Peptides/physiology , Peptides/toxicity , Polyketides/toxicity , Probiotics/toxicity , Protein Domains , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/therapy , Salmonella typhimurium , Siderophores/genetics , Siderophores/physiology , Siderophores/toxicity , Virulence Factors/genetics , Virulence Factors/physiology , Virulence Factors/toxicity
12.
Mol Cell ; 74(2): 227-229, 2019 04 18.
Article En | MEDLINE | ID: mdl-31002804

In a recent issue of Science, Wilson et al. (2019) provide direct evidence that the bacterial-produced colibactin alkylates DNA in vivo, resulting in DNA adducts, which mediates its genotoxic effect. This work reinforces the role of colibactin-producing bacteria in colon cancer pathogenesis.


Colorectal Neoplasms/microbiology , Escherichia coli/genetics , Gastrointestinal Microbiome/genetics , Peptides/toxicity , Polyketides/toxicity , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Adducts/genetics , DNA Adducts/toxicity , DNA Damage/drug effects , Escherichia coli/pathogenicity , Humans , Mutagens/metabolism , Mutagens/toxicity , Mutation/drug effects , Mutation/genetics , Peptides/genetics
13.
Science ; 363(6428)2019 02 15.
Article En | MEDLINE | ID: mdl-30765538

Certain Escherichia coli strains residing in the human gut produce colibactin, a small-molecule genotoxin implicated in colorectal cancer pathogenesis. However, colibactin's chemical structure and the molecular mechanism underlying its genotoxic effects have remained unknown for more than a decade. Here we combine an untargeted DNA adductomics approach with chemical synthesis to identify and characterize a covalent DNA modification from human cell lines treated with colibactin-producing E. coli Our data establish that colibactin alkylates DNA with an unusual electrophilic cyclopropane. We show that this metabolite is formed in mice colonized by colibactin-producing E. coli and is likely derived from an initially formed, unstable colibactin-DNA adduct. Our findings reveal a potential biomarker for colibactin exposure and provide mechanistic insights into how a gut microbe may contribute to colorectal carcinogenesis.


Carcinogenesis/metabolism , Colorectal Neoplasms/microbiology , Cyclopropanes/metabolism , DNA Adducts/metabolism , DNA Damage , Escherichia coli/metabolism , Gastrointestinal Microbiome , Mutagens/metabolism , Peptides/metabolism , Polyketides/metabolism , Alkylating Agents , Alkylation , Animals , Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Cyclopropanes/chemistry , Escherichia coli/pathogenicity , Germ-Free Life , HT29 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mutagens/toxicity , Peptides/chemistry , Peptides/toxicity , Polyketides/chemistry , Polyketides/toxicity
14.
Toxins (Basel) ; 10(4)2018 04 10.
Article En | MEDLINE | ID: mdl-29642622

Cyclomodulins are bacterial toxins that interfere with the eukaryotic cell cycle. A new cyclomodulin called colibactin, which is synthetized by the pks genomic island, was discovered in 2006. Despite many efforts, colibactin has not yet been purified, and its structure remains elusive. Interestingly, the pks island is found in members of the family Enterobacteriaceae (mainly Escherichia coli and Klebsiella pneumoniae) isolated from different origins, including from intestinal microbiota, septicaemia, newborn meningitis, and urinary tract infections. Colibactin-producing bacteria induce chromosomal instability and DNA damage in eukaryotic cells, which leads to senescence of epithelial cells and apoptosis of immune cells. The pks island is mainly observed in B2 phylogroup E. coli strains, which include extra-intestinal pathogenic E. coli strains, and pksE. coli are over-represented in biopsies isolated from colorectal cancer. In addition, pksE. coli bacteria increase the number of tumours in diverse colorectal cancer mouse models. Thus, colibactin could have a major impact on human health. In the present review, we will focus on the biological effects of colibactin, the distribution of the pks island, and summarize what is currently known about its synthesis and its structure.


Bacterial Toxins , Mutagens , Peptides , Polyketides , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Genomic Islands , Humans , Mutagens/chemistry , Mutagens/metabolism , Mutagens/toxicity , Peptides/chemistry , Peptides/metabolism , Peptides/toxicity , Polyketides/chemistry , Polyketides/metabolism , Polyketides/toxicity
15.
Z Naturforsch C J Biosci ; 73(3-4): 161-163, 2018 Feb 23.
Article En | MEDLINE | ID: mdl-29320367

Two compounds, compounds 1 and 2, were isolated from Preussia sp. The molecular structures of both compounds were elucidated by analyzing one-dimensional (1D) and 2D nuclear magnetic resonance data along with high-resolution mass spectrometry data. Compound 1 was obtained as novel in structure, and compound 2 was recently reported elsewhere. Compound 1 did not show antioxidant and antimicrobial activities and brine shrimp toxicity, while compound 2 showed strong antioxidant activity (DPPH reduction capacity; IC50=3 µg/mL) and brine shrimp toxicity (LD50=50 µg/mL).


Anti-Infective Agents/chemistry , Antioxidants/chemistry , Ascomycota/chemistry , Lichens/chemistry , Polyketides/chemistry , Anti-Infective Agents/toxicity , Antioxidants/toxicity , Polyketides/toxicity , Staphylococcus aureus/drug effects
16.
J Am Chem Soc ; 139(49): 17719-17722, 2017 12 13.
Article En | MEDLINE | ID: mdl-29112397

Certain commensal Escherichia coli contain the clb biosynthetic gene cluster that codes for small molecule prodrugs known as precolibactins. Precolibactins are converted to colibactins by N-deacylation; the latter are postulated to be genotoxic and to contribute to colorectal cancer formation. Though advances toward elucidating (pre)colibactin biosynthesis have been made, the functions and mechanisms of several clb gene products remain poorly understood. Here we report the 2.1 Å X-ray structure and molecular function of ClbS, a gene product that confers resistance to colibactin toxicity in host bacteria and which has been shown to be important for bacterial viability. The structure harbors a potential colibactin binding site and shares similarity to known hydrolases. In vitro studies using a synthetic colibactin analog and ClbS or an active site residue mutant reveal cyclopropane hydrolase activity that converts the electrophilic cyclopropane of the colibactins into an innocuous hydrolysis product. As the cyclopropane has been shown to be essential for genotoxic effects in vitro, this ClbS-catalyzed ring-opening provides a means for the bacteria to circumvent self-induced genotoxicity. Our study provides a molecular-level view of the first reported cyclopropane hydrolase and support for a specific mechanistic role of this enzyme in colibactin resistance.


Cyclopropanes/metabolism , Drug Resistance , Escherichia coli/enzymology , Escherichia coli/metabolism , Hydrolases/metabolism , Peptides/metabolism , Polyketides/metabolism , Binding Sites , Crystallography, X-Ray , Cyclopropanes/chemistry , Drug Resistance/drug effects , Escherichia coli/drug effects , Escherichia coli/genetics , Hydrolases/chemistry , Microbial Viability/drug effects , Peptides/chemistry , Peptides/pharmacology , Peptides/toxicity , Polyketides/chemistry , Polyketides/pharmacology , Polyketides/toxicity
18.
Pathog Dis ; 75(5)2017 07 31.
Article En | MEDLINE | ID: mdl-28637194

The genotoxin colibactin is a secondary metabolite produced by a variety of pathogenic Enterobacteria, and is associated with colon cancer development and acute systemic infections. The colibactin biosynthesis requires the enzymatic activity of the phosphopantetheinyl transferase ClbA. We recently evidenced that two master regulators of bacterial iron homeostasis, i.e. the ferric uptake regulator (Fur) and the small regulatory non-coding RNA RyhB, were involved in the regulation of the clbA transcription and of the colibactin production. In this study, we investigated the impact of high iron supply on clbA transcription and colibactin production in wild type, ΔryhB, Δfur and ΔryhB Δfur strains. This revealed that high iron resulted in decreased synthesis of the genotoxin colibactin through both pathways dependent and independent of Fur/RyhB. This work highlights the complex regulatory mechanism that controls an important bacterial virulence and carcinogenesis factor by regulators of bacterial iron homeostasis.


Bacterial Proteins/genetics , Escherichia coli/drug effects , Gene Expression Regulation, Bacterial , Iron/metabolism , Mutagens/metabolism , Peptides/genetics , RNA, Small Untranslated/genetics , Repressor Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/pathogenicity , Gene Deletion , HeLa Cells , Humans , Iron/pharmacology , Mutagens/chemistry , Mutagens/toxicity , Peptides/antagonists & inhibitors , Peptides/metabolism , Peptides/toxicity , Polyketides/antagonists & inhibitors , Polyketides/metabolism , Polyketides/toxicity , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Small Untranslated/metabolism , Repressor Proteins/deficiency , Signal Transduction , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription, Genetic , Transferases (Other Substituted Phosphate Groups)/genetics , Transferases (Other Substituted Phosphate Groups)/metabolism , Virulence
19.
Microb Pathog ; 110: 117-127, 2017 Sep.
Article En | MEDLINE | ID: mdl-28652178

The chromanequinone (BIQ) compound produced by the mangrove estuary derived strain, Streptomyces sp. JRG-04 was effective even at low MIC level concentration against Methicillin resistant S. aureus and other clinical pathogens. In this study, we have investigated the antimicrobial potential of chromanequinone compound by using various microscopy and imaging techniques. The flow cytometry (FACS) analysis suggested the BIQ aromatic polyketide compound produced by the Streptomyces sp. JRG-04 has toxic effect on MRSA cell membrane by increased up take of propidium iodide dye. The bacterial imaging analysis by high content screening experiment (HCS) revealed the increased number of dead MRSA cells than the live MRSA populations with chromanequinone treatment. Furthermore, atomic force microscopic study proved the MRSA cell surface ultra-structure changes when the cells exposed to chromanequinone compound at 3 h and 6 h. Further, in-vitro lymphocytotoxicity effect of chromanequinone compound at different concentrations with the combination of complement was performed on human lymphocytes by cell lysis assay. Interestingly, we have found that the higher concentration of BIQ chromanequinone (10 mg/mL) compound without complement induced apoptosis of human lymphocytes. The present investigation reveals that the toxic potential of chromanequinone on human lymphocytes might be associated with the complement dependent. This study strongly suggests that the chromanequinone compound produced by the Streptomyces strain with bioactive property can be developed as a therapeutic leads for various pharmaceutical applications.


Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Polyketides/pharmacology , Polyketides/toxicity , Streptomyces/metabolism , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Erythrocytes/drug effects , Flow Cytometry , Humans , Lymphocytes/drug effects , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Microbial Sensitivity Tests
20.
mBio ; 8(2)2017 03 14.
Article En | MEDLINE | ID: mdl-28292979

An increasing number of human beings from developed countries are colonized by Escherichia coli strains producing colibactin, a genotoxin suspected to be associated with the development of colorectal cancers. Deoxynivalenol (DON) is the most prevalent mycotoxin that contaminates staple food-especially cereal products-in Europe and North America. This study investigates the effect of the food contaminant DON on the genotoxicity of the E. coli strains producing colibactin. In vitro, intestinal epithelial cells were coexposed to DON and E. coli producing colibactin. In vivo, newborn rats colonized at birth with E. coli producing colibactin were fed a DON-contaminated diet. Intestinal DNA damage was estimated by the phosphorylation of histone H2AX. DON exacerbates the genotoxicity of the E. coli producing colibactin in a time- and dose-dependent manner in vitro Although DON had no effect on the composition of the gut microbiota, and especially on the number of E. coli, a significant increase in DNA damage was observed in intestinal epithelial cells of animals colonized by E. coli strains producing colibactin and coexposed to DON compared to animals colonized with E. coli strains unable to produce colibactin or animals exposed only to DON. In conclusion, our data demonstrate that the genotoxicity of E. coli strains producing colibactin, increasingly present in the microbiota of asymptomatic human beings, is modulated by the presence of DON in the diet. This raises questions about the synergism between food contaminants and gut microbiota with regard to intestinal carcinogenesis.IMPORTANCE An increasing number of human beings from developed countries are colonized by Escherichia coli strains producing colibactin, a genotoxin suspected to be associated with the development of colorectal cancers. Deoxynivalenol (DON) is the most prevalent mycotoxin that contaminates staple food-especially cereal products-in Europe and North America. Our in vitro and in vivo results demonstrate that the intestinal DNA damage induced by colibactin-producing E. coli strains was exacerbated by the presence of DON in the diet. This raises questions about the synergism between food contaminants and gut microbiota with regard to intestinal carcinogenesis.


DNA Damage/drug effects , Escherichia coli/drug effects , Escherichia coli/metabolism , Gastrointestinal Tract/microbiology , Mutagens/toxicity , Peptides/toxicity , Polyketides/toxicity , Trichothecenes/metabolism , Animals , Coculture Techniques , Epithelial Cells/drug effects , Escherichia coli/growth & development , Histones/analysis , Rats
...