Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.664
Filtrar
1.
Gut Microbes ; 16(1): 2397874, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39229962

RESUMEN

Recently, the mounting integration of probiotics into human health strategies has gathered considerable attention. Although the benefits of probiotics have been widely recognized in patients with gastrointestinal disorders, immune system modulation, and chronic-degenerative diseases, there is a growing need to evaluate their potential risks. In this context, new concerns have arisen regarding the safety of probiotics as some strains may have adverse effects in humans. Among these strains, Escherichia coli Nissle 1917 (EcN) exhibited traits of concern due to a pathogenic locus in its genome that produces potentially genotoxic metabolites. As the use of probiotics for therapeutic purposes is increasing, the effects of potentially harmful probiotics must be carefully evaluated. To this end, in this narrative review article, we reported the findings of the most relevant in vitro and in vivo studies investigating the expanding applications of probiotics and their impact on human well-being addressing concerns arising from the presence of antibiotic resistance and pathogenic elements, with a focus on the polyketide synthase (pks) pathogenic island of EcN. In this context, the literature data here discussed encourages a thorough profiling of probiotics to identify potential harmful elements as done for EcN where potential genotoxic effects of colibactin, a secondary metabolite, were observed. Specifically, while some studies suggest EcN is safe for gastrointestinal health, conflicting findings highlight the need for further research to clarify its safety and optimize its use in therapy. Overall, the data here presented suggest that a comprehensive assessment of the evolving landscape of probiotics is essential to make evidence-based decisions and ensure their correct use in humans.


Asunto(s)
Escherichia coli , Péptidos , Policétidos , Probióticos , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Policétidos/metabolismo , Péptidos/metabolismo , Péptidos/genética , Animales , Mutágenos/metabolismo , Mutágenos/toxicidad , Daño del ADN , Sintasas Poliquetidas/genética , Sintasas Poliquetidas/metabolismo
2.
Chem Res Toxicol ; 37(8): 1364-1373, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-38900731

RESUMEN

Chemicals often require metabolic activation to become genotoxic. Established test guidelines recommend the use of the rat liver S9 fraction or microsomes to introduce metabolic competence to in vitro cell-based bioassays, but the use of animal-derived components in cell culture raises ethical concerns and may lead to quality issues and reproducibility problems. The aim of the present study was to compare the metabolic activation of cyclophosphamide (CPA) and benzo[a]pyrene (BaP) by induced rat liver microsomes and an abiotic cytochrome P450 (CYP) enzyme based on a biomimetic porphyrine catalyst. For the detection of genotoxic effects, the chemicals were tested in a reporter gene assay targeting the activation of the cellular tumor protein p53. Both chemicals were metabolized by the abiotic CYP enzyme and the microsomes. CPA showed no activation of p53 and low cytotoxicity without metabolic activation, but strong activation of p53 and increased cytotoxicity upon incubation with liver microsomes or abiotic CYP enzyme. The effect concentration causing a 1.5-fold induction of p53 activation was very similar with both metabolization systems (within a factor of 1.5), indicating that genotoxic metabolites were formed at comparable concentrations. BaP also showed low cytotoxicity and no p53 activation without metabolic activation. The activation of p53 was detected for BaP upon incubation with active and inactive microsomes at similar concentrations, indicating experimental artifacts caused by the microsomes or NADPH. The activation of BaP with the abiotic CYP enzyme increased the cytotoxicity of BaP by a factor of 8, but no activation of p53 was detected. The results indicate that abiotic CYP enzymes may present an alternative to rat liver S9 fraction or microsomes for the metabolic activation of test chemicals, which are completely free of animal-derived components. However, an amendment of existing test guidelines would require testing of more chemicals and genotoxicity end points.


Asunto(s)
Benzo(a)pireno , Sistema Enzimático del Citocromo P-450 , Microsomas Hepáticos , Proteína p53 Supresora de Tumor , Microsomas Hepáticos/metabolismo , Animales , Ratas , Benzo(a)pireno/metabolismo , Benzo(a)pireno/toxicidad , Benzo(a)pireno/química , Sistema Enzimático del Citocromo P-450/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ciclofosfamida/metabolismo , Ciclofosfamida/toxicidad , Mutágenos/toxicidad , Mutágenos/metabolismo , Mutágenos/química , Masculino , Activación Metabólica , Humanos , Supervivencia Celular/efectos de los fármacos
3.
Mycotoxin Res ; 40(3): 331-346, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38587710

RESUMEN

Zearalenone (ZEN) and deoxynivalenol (DON) and their derivatives are well-known mycotoxins, which can occur not only in crops but also in water bodies, including drinking water sources. In vitro bioassays can be used to detect biological effects of hazardous compounds in water. To this, when studying biological effects and toxicity in vitro, metabolism is important to consider. In this study, ZEN, α-zearalenol (α-ZEL), DON, 3-acetyl DON, and 15-acetyl DON were evaluated in vitro for hormone receptor-mediated effects (estrogen receptor [ER] and androgen receptor [AR]) and genotoxicity (micronucleus assay) in the presence of an exogenous metabolic activation system (MAS). The ER bioassay proved to be a highly sensitive method to detect low concentrations of the ZEN compounds (EC10 values of 31.4 pM for ZEN, 3.59 pM for α-ZEL) in aqueous solutions. In the presence of the MAS, reduced estrogenic effects were observed for both ZEN compounds (EC10 values of 6.47 × 103 pM for ZEN, 1.55 × 102 pM for α-ZEL). Of the DON compounds, only 3-acetyl DON was estrogenic (EC10 of 0.31 µM), and the effect was removed in the presence of the MAS. Anti-androgenic effects of the ZEN compounds and androgenic effects of the DON compounds were detected in the micromolar range. No induction of genotoxicity was detected for ZEN or DON in the presence of the MAS. Our study highlighted that inclusion of exogenous MAS is a useful tool to detect biological effects of metabolites in in vitro bioassays.


Asunto(s)
Bioensayo , Tricotecenos , Zearalenona , Zearalenona/toxicidad , Tricotecenos/toxicidad , Tricotecenos/metabolismo , Bioensayo/métodos , Receptores Androgénicos/metabolismo , Andrógenos/metabolismo , Andrógenos/toxicidad , Humanos , Mutágenos/toxicidad , Mutágenos/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/efectos de los fármacos , Estrógenos/metabolismo , Estrógenos/toxicidad , Pruebas de Micronúcleos
4.
Gut Microbes ; 16(1): 2310215, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38374654

RESUMEN

Human colorectal cancers (CRCs) are readily colonized by colibactin-producing E. coli (CoPEC). CoPEC induces DNA double-strand breaks, DNA mutations, genomic instability, and cellular senescence. Infected cells produce a senescence-associated secretory phenotype (SASP), which is involved in the increase in tumorigenesis observed in CRC mouse models infected with CoPEC. This study investigated whether CoPEC, and the SASP derived from CoPEC-infected cells, impacted chemotherapeutic resistance. Human intestinal epithelial cells were infected with the CoPEC clinical 11G5 strain or with its isogenic mutant, which is unable to produce colibactin. Chemotherapeutic resistance was assessed in vitro and in a xenograft mouse model. Expressions of cancer stem cell (CSC) markers in infected cells were investigated. Data were validated using a CRC mouse model and human clinical samples. Both 11G5-infected cells, and uninfected cells incubated with the SASP produced by 11G5-infected cells exhibited an increased resistance to chemotherapeutic drugs in vitro and in vivo. This finding correlated with the induction of the epithelial to mesenchymal transition (EMT), which led to the emergence of cells exhibiting CSC features. They grew on ultra-low attachment plates, formed colonies in soft agar, and overexpressed several CSC markers (e.g. CD133, OCT-3/4, and NANOG). In agreement with these results, murine and human CRC biopsies colonized with CoPEC exhibited higher expression levels of OCT-3/4 and NANOG than biopsies devoid of CoPEC. Conclusion: CoPEC might aggravate CRCs by inducing the emergence of cancer stem cells that are highly resistant to chemotherapy.


Asunto(s)
Microbioma Gastrointestinal , Neoplasias , Péptidos , Policétidos , Humanos , Ratones , Animales , Escherichia coli/genética , Escherichia coli/metabolismo , Transición Epitelial-Mesenquimal , Mutágenos/metabolismo , Policétidos/farmacología , Policétidos/metabolismo , Modelos Animales de Enfermedad , Células Madre Neoplásicas/metabolismo
5.
Gut Microbes ; 16(1): 2320291, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38417029

RESUMEN

Intratumoral bacteria flexibly contribute to cellular and molecular tumor heterogeneity for supporting cancer recurrence through poorly understood mechanisms. Using spatial metabolomic profiling technologies and 16SrRNA sequencing, we herein report that right-sided colorectal tumors are predominantly populated with Colibactin-producing Escherichia coli (CoPEC) that are locally establishing a high-glycerophospholipid microenvironment with lowered immunogenicity. It coincided with a reduced infiltration of CD8+ T lymphocytes that produce the cytotoxic cytokines IFN-γ where invading bacteria have been geolocated. Mechanistically, the accumulation of lipid droplets in infected cancer cells relied on the production of colibactin as a measure to limit genotoxic stress to some extent. Such heightened phosphatidylcholine remodeling by the enzyme of the Land's cycle supplied CoPEC-infected cancer cells with sufficient energy for sustaining cell survival in response to chemotherapies. This accords with the lowered overall survival of colorectal patients at stage III-IV who were colonized by CoPEC when compared to patients at stage I-II. Accordingly, the sensitivity of CoPEC-infected cancer cells to chemotherapies was restored upon treatment with an acyl-CoA synthetase inhibitor. By contrast, such metabolic dysregulation leading to chemoresistance was not observed in human colon cancer cells that were infected with the mutant strain that did not produce colibactin (11G5∆ClbQ). This work revealed that CoPEC locally supports an energy trade-off lipid overload within tumors for lowering tumor immunogenicity. This may pave the way for improving chemoresistance and subsequently outcome of CRC patients who are colonized by CoPEC.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Péptidos , Policétidos , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Microambiente Tumoral , Resistencia a Antineoplásicos , Mutágenos/metabolismo , Recurrencia Local de Neoplasia , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/microbiología , Policétidos/metabolismo , Lípidos
6.
J Toxicol Sci ; 48(11): 571-583, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37914285

RESUMEN

Various chemicals, including pesticides, heavy metals, and metabolites of tobacco, have been detected in fetal environment. Fetuses are exposed to these chemicals at relatively low concentrations; however, their risk of developing neurological and behavioral disorders increases after birth. We aimed to evaluate the effects of five chemicals (diethylphosphate, cotinine, octachlorodipropyl ether, mercury, and selenium) detected in the serum of pregnant mothers on neural development using human neurospheres (NSphs) differentiated from induced pluripotent stem cells. Exposure to each chemical at serum concentrations revealed no effects on NSph development. However, combined exposure to the five chemicals caused a significant decrease in NSph size and altered gene expression and neural differentiation. Thus, we next focused on DNA methylation to investigate changes in NSph properties caused by chemical exposure. Combined exposure to chemicals had extremely small effects on the DNA methylation status of NSphs at individual gene loci. However, stochastic changes in methylation status caused by chemical exposure were significantly accumulated throughout the entire genome. These results suggest that the five chemicals acted as epimutagens that alter the epigenetic status during human neural development at the biological level. Taken together, we showed for the first time, the epimutagen-induced alterations in neural differentiation at serum concentrations using an in vitro human neuronal model.


Asunto(s)
Epigénesis Genética , Células Madre Pluripotentes Inducidas , Embarazo , Femenino , Humanos , Mutágenos/metabolismo , Metilación de ADN , Diferenciación Celular/genética
7.
Neoplasia ; 43: 100918, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37499275

RESUMEN

Certain Enterobacteriaceae strains contain a 54-kb biosynthetic gene cluster referred to as "pks" encoding the biosynthesis of a secondary metabolite, colibactin. Colibactin-producing E. coli promote colorectal cancer (CRC) in preclinical models, and in vitro induce a specific mutational signature that is also detected in human CRC genomes. Yet, how colibactin exposure affects the mutational landscape of CRC in vivo remains unclear. Here we show that colibactin-producing E. coli-driven colonic tumors in mice have a significantly higher SBS burden and a larger percentage of these mutations can be attributed to a signature associated with mismatch repair deficiency (MMRd; SBS15), compared to tumors developed in the presence of colibactin-deficient E. coli. We found that the synthetic colibactin 742 but not an inactive analog 746 causes DNA damage and induces transcriptional activation of p53 and senescence signaling pathways in non-transformed human colonic epithelial cells. In MMRd colon cancer cells (HCT 116), chronic exposure to 742 resulted in the upregulation of BRCA1, Fanconi anemia, and MMR signaling pathways as revealed by global transcriptomic analysis. This was accompanied by increased T>N single-base substitutions (SBS) attributed to the proposed pks+E. coli signature (SBS88), reactive oxygen species (SBS17), and mismatch-repair deficiency (SBS44). A significant co-occurrence between MMRd SBS44 and pks-associated SBS88 signature was observed in a large cohort of human CRC patients (n=2,945), and significantly more SBS44 mutations were found when SBS88 was also detected. Collectively, these findings reveal the host response mechanisms underlying colibactin genotoxic activity and suggest that colibactin may exacerbate MMRd-associated mutations.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Humanos , Ratones , Animales , Mutágenos/toxicidad , Mutágenos/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Reparación de la Incompatibilidad de ADN/genética , Mutación , Neoplasias Colorrectales/genética , Neoplasias del Colon/patología
9.
Nat Chem Biol ; 19(2): 159-167, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36253549

RESUMEN

The human gut bacterial genotoxin colibactin is a possible key driver of colorectal cancer (CRC) development. Understanding colibactin's biological effects remains difficult owing to the instability of the proposed active species and the complexity of the gut microbiota. Here, we report small molecule boronic acid inhibitors of colibactin biosynthesis. Designed to mimic the biosynthetic precursor precolibactin, these compounds potently inhibit the colibactin-activating peptidase ClbP. Using biochemical assays and crystallography, we show that they engage the ClbP binding pocket, forming a covalent bond with the catalytic serine. These inhibitors reproduce the phenotypes observed in a clbP deletion mutant and block the genotoxic effects of colibactin on eukaryotic cells. The availability of ClbP inhibitors will allow precise, temporal control over colibactin production, enabling further study of its contributions to CRC. Finally, application of our inhibitors to related peptidase-encoding pathways highlights the power of chemical tools to probe natural product biosynthesis.


Asunto(s)
Microbioma Gastrointestinal , Policétidos , Humanos , Mutágenos/metabolismo , Mutágenos/toxicidad , Escherichia coli/metabolismo , Policétidos/química , Péptido Hidrolasas/química
10.
Science ; 378(6618): 358-359, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36302018
11.
Science ; 378(6618): eabm3233, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36302024

RESUMEN

Microbiota-derived metabolites that elicit DNA damage can contribute to colorectal cancer (CRC). However, the full spectrum of genotoxic chemicals produced by indigenous gut microbes remains to be defined. We established a pipeline to systematically evaluate the genotoxicity of an extensive collection of gut commensals from inflammatory bowel disease patients. We identified isolates from divergent phylogenies whose metabolites caused DNA damage and discovered a distinctive family of genotoxins-termed the indolimines-produced by the CRC-associated species Morganella morganii. A non-indolimine-producing M. morganii mutant lacked genotoxicity and failed to exacerbate colon tumorigenesis in mice. These studies reveal the existence of a previously unexplored universe of genotoxic small molecules from the microbiome that may affect host biology in homeostasis and disease.


Asunto(s)
Neoplasias Colorrectales , Daño del ADN , Microbioma Gastrointestinal , Indoles , Enfermedades Inflamatorias del Intestino , Morganella morganii , Mutágenos , Animales , Ratones , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/microbiología , Enfermedades Inflamatorias del Intestino/microbiología , Morganella morganii/genética , Morganella morganii/aislamiento & purificación , Morganella morganii/metabolismo , Indoles/metabolismo , Carcinogénesis/genética , Humanos , Mutágenos/metabolismo , Células HeLa
12.
PLoS Pathog ; 18(9): e1010766, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36067266

RESUMEN

Wound infections are often polymicrobial in nature, biofilm associated and therefore tolerant to antibiotic therapy, and associated with delayed healing. Escherichia coli and Staphylococcus aureus are among the most frequently cultured pathogens from wound infections. However, little is known about the frequency or consequence of E. coli and S. aureus polymicrobial interactions during wound infections. Here we show that E. coli kills Staphylococci, including S. aureus, both in vitro and in a mouse excisional wound model via the genotoxin, colibactin. Colibactin biosynthesis is encoded by the pks locus, which we identified in nearly 30% of human E. coli wound infection isolates. While it is not clear how colibactin is released from E. coli or how it penetrates target cells, we found that the colibactin intermediate N-myristoyl-D-Asn (NMDA) disrupts the S. aureus membrane. We also show that the BarA-UvrY two component system (TCS) senses the environment created during E. coli and S. aureus mixed species interaction, leading to upregulation of pks island genes. Further, we show that BarA-UvrY acts via the carbon storage global regulatory (Csr) system to control pks expression. Together, our data demonstrate the role of colibactin in interspecies competition and show that it is regulated by BarA-UvrY TCS during interspecies competition.


Asunto(s)
Infecciones por Escherichia coli , Proteínas de Escherichia coli , Proteínas de la Membrana , Fosfotransferasas , Policétidos , Staphylococcus aureus , Factores de Transcripción , Animales , Antibacterianos/metabolismo , Carbono/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Mutágenos/metabolismo , N-Metilaspartato/metabolismo , Péptidos , Fosfotransferasas/genética , Policétidos/metabolismo , Staphylococcus/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Factores de Transcripción/metabolismo , Infección de Heridas/microbiología
13.
Chem Res Toxicol ; 35(10): 1747-1765, 2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-36044734

RESUMEN

Nitro group containing xenobiotics include drugs, cancer chemotherapeutic agents, carcinogens (e.g., nitroarenes and aristolochic acid) and explosives. The nitro group undergoes a six-electron reduction to form sequentially the nitroso-, N-hydroxylamino- and amino-functional groups. These reactions are catalyzed by nitroreductases which, rather than being enzymes with this sole function, are enzymes hijacked for their propensity to donate electrons to the nitro group either one at a time via a radical mechanism or two at time via the equivalent of a hydride transfer. These enzymes include: NADPH-dependent flavoenzymes (NADPH: P450 oxidoreductase, NAD(P)H-quinone oxidoreductase), P450 enzymes, oxidases (aldehyde oxidase, xanthine oxidase) and aldo-keto reductases. The hydroxylamino group once formed can undergo conjugation reactions with acetate or sulfate catalyzed by N-acetyltransferases or sulfotransferases, respectively, leading to the formation of intermediates containing a good leaving group which in turn can generate a nitrenium or carbenium ion for covalent DNA adduct formation. The intermediates in the reduction sequence are also prone to oxidation and produce reactive oxygen species. As a consequence, many nitro-containing xenobiotics can be genotoxic either by forming stable covalent adducts or by oxidatively damaging DNA. This review will focus on the general chemistry of nitroreduction, the enzymes responsible, the reduction of xenobiotic substrates, the regulation of nitroreductases, the ability of nitrocompounds to form DNA adducts and act as mutagens as well as some future directions.


Asunto(s)
Contaminantes Ambientales , Sustancias Explosivas , Acetiltransferasas/metabolismo , Aldehídos , Aldo-Ceto Reductasas/metabolismo , Carcinógenos , Aductos de ADN , Redes y Vías Metabólicas , Mutágenos/metabolismo , NAD/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , NADP/metabolismo , Quinonas , Especies Reactivas de Oxígeno , Sulfatos , Sulfotransferasas/metabolismo , Xantina Oxidasa/metabolismo , Xenobióticos
14.
Virulence ; 13(1): 1199-1215, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35795909

RESUMEN

Bacterial genotoxins are peptide or protein virulence factors produced by several pathogens, which make single-strand breaks (SSBs) and/or double-strand DNA breaks (DSBs) in the target host cells. If host DNA inflictions are not resolved on time, host cell apoptosis, cell senescence, and/or even bacterial pathogen-related cancer may occur. Two multi-protein AB toxins, cytolethal distending toxin (CDT) produced by over 30 bacterial pathogens and typhoid toxin from Salmonella Typhi, as well as small polyketide-peptides named colibactin that causes the DNA interstrand cross-linking and subsequent DSBs is the most well-characterized bacterial genotoxins. Using these three examples, this review discusses the mechanisms by which these toxins deliver themselves into the nucleus of the target host cells and exert their genotoxic functions at the structural and functional levels.


Asunto(s)
Bacterias , Mutágenos , Bacterias/metabolismo , Roturas del ADN de Doble Cadena , Daño del ADN , Mutágenos/metabolismo , Mutágenos/toxicidad , Factores de Virulencia/genética
15.
Artículo en Inglés | MEDLINE | ID: mdl-35483777

RESUMEN

Sodium azide is a strong mutagen that has been successfully employed in mutation breeding of crop plants. In biological systems, it is metabolically converted to the proximate mutagen azidoalanine, which requires further bioactivation to a putative ultimate mutagen that remains elusive. The nature of the DNA modifications induced by azides leading to mutations is also unknown. Other mutagenic organic azido compounds seem to share the same bioactivation pathway to the ultimate mutagenic species as they induce point mutations dependent on the same DNA repair pathways. We investigated mutations induced by the representative mutagen 3-azido-1,2-propanediol (azidoglycerol, AZG) in the human TK6 cell line. Until now, azides have been considered to be non-mutagens and non-carcinogens in mammals, including humans, as judged only by the conventional clastogenicity chromosomal aberration types of bioassays. Here, we show the potent mutagenicity of AZG in cultured human cells, comparable to alkylating agents such as methyl methanesulfonate at concentrations with similar lethality. The potent ability of an organic azide to induce base substitutions in a mammalian system raises an alert with respect to human exposure to organic and inorganic azido compounds.


Asunto(s)
Azidas , Mutágenos , Animales , Azidas/metabolismo , Azidas/toxicidad , Humanos , Mamíferos , Mutagénesis , Pruebas de Mutagenicidad , Mutágenos/metabolismo , Mutágenos/toxicidad , Glicoles de Propileno
16.
Nat Prod Rep ; 39(5): 991-1014, 2022 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-35288725

RESUMEN

Covering: up to 2021Colibactin(s), a group of secondary metabolites produced by the pks island (clb cluster) of Escherichia coli, shows genotoxicity relevant to colorectal cancer and thus significantly affects human health. Over the last 15 years, substantial efforts have been exerted to reveal the molecular structure of colibactin, but progress is slow owing to its instability, low titer, and elusive and complex biosynthesis logic. Fortunately, benefiting from the discovery of the prodrug mechanism, over 40 precursors of colibactin have been reported. Some key biosynthesis genes located on the pks island have also been characterised. Using an integrated bioinformatics, metabolomics, and chemical synthesis approach, researchers have recently characterised the structure and possible biosynthesis processes of colibactin, thereby providing new insights into the unique biosynthesis logic and the underlying mechanism of the biological activity of colibactin. Early developments in the study of colibactin have been summarised in several previous reviews covering various study periods, whereas the two most recent reviews have focused primarily on the chemical synthesis of colibactin. The present review aims to provide an update on the biosynthesis and bioactivities of colibactin.


Asunto(s)
Mutágenos , Policétidos , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Mutágenos/metabolismo , Péptidos/química , Policétidos/metabolismo
17.
Proc Natl Acad Sci U S A ; 119(11): e2121180119, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35254905

RESUMEN

SignificanceIn a polymicrobial battlefield where different species compete for nutrients and colonization niches, antimicrobial compounds are the sword and shield of commensal microbes in competition with invading pathogens and each other. The identification of an Escherichia coli-produced genotoxin, colibactin, and its specific targeted killing of enteric pathogens and commensals, including Vibrio cholerae and Bacteroides fragilis, sheds light on our understanding of intermicrobial interactions in the mammalian gut. Our findings elucidate the mechanisms through which genotoxins shape microbial communities and provide a platform for probing the larger role of enteric multibacterial interactions regarding infection and disease outcomes.


Asunto(s)
Cólera/microbiología , Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Interacciones Microbianas , Mutágenos/metabolismo , Vibrio cholerae/fisiología , Animales , Antibiosis , Cólera/mortalidad , Daño del ADN , Modelos Animales de Enfermedad , Escherichia coli/fisiología , Humanos , Ratones , Péptidos/metabolismo , Péptidos/farmacología , Policétidos/metabolismo , Policétidos/farmacología , Pronóstico , Especies Reactivas de Oxígeno , Vibrio cholerae/efectos de los fármacos
18.
Environ Sci Pollut Res Int ; 29(41): 62111-62159, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34611806

RESUMEN

The etiology of the majority of human cancers is associated with a myriad of environmental causes, including physical, chemical, and biological factors. DNA damage induced by such mutagens is the initial step in the process of carcinogenesis resulting in the accumulation of mutations. Mutational events are considered the major triggers for introducing genetic and epigenetic insults such as DNA crosslinks, single- and double-strand DNA breaks, formation of DNA adducts, mismatched bases, modification in histones, DNA methylation, and microRNA alterations. However, DNA repair mechanisms are devoted to protect the DNA to ensure genetic stability, any aberrations in these calibrated mechanisms provoke cancer occurrence. Comprehensive knowledge of the type of mutagens and carcinogens and the influence of these agents in DNA damage and cancer induction is crucial to develop rational anticancer strategies. This review delineated the molecular mechanism of DNA damage and the repair pathways to provide a deep understanding of the molecular basis of mutagenicity and carcinogenicity. A relationship between DNA adduct formation and cancer incidence has also been summarized. The mechanistic basis of inflammatory response and oxidative damage triggered by mutagens in tumorigenesis has also been highlighted. We elucidated the interesting interplay between DNA damage response and immune system mechanisms. We addressed the current understanding of DNA repair targeted therapies and DNA damaging chemotherapeutic agents for cancer treatment and discussed how antiviral agents, anti-inflammatory drugs, and immunotherapeutic agents combined with traditional approaches lay the foundations for future cancer therapies.


Asunto(s)
Mutágenos , Neoplasias , Carcinógenos/metabolismo , Carcinógenos/toxicidad , ADN/metabolismo , Daño del ADN , Reparación del ADN , Humanos , Mutágenos/metabolismo , Mutágenos/toxicidad , Neoplasias/epidemiología
19.
J Am Chem Soc ; 143(38): 15824-15833, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34524796

RESUMEN

Colibactin is a genotoxic metabolite produced by commensal-pathogenic members of the human microbiome that possess the clb (aka pks) biosynthetic gene cluster. clb+ bacteria induce tumorigenesis in models of intestinal inflammation and have been causally linked to oncogenesis in humans. While colibactin is believed underlie these effects, it has not been possible to study the molecule directly due to its instability. Herein, we report the synthesis and biological studies of colibactin 742 (4), a stable colibactin derivative. We show that colibactin 742 (4) induces DNA interstrand-cross-links, activation of the Fanconi Anemia DNA repair pathway, and G2/M arrest in a manner similar to clb+E. coli. The linear precursor 9, which mimics the biosynthetic precursor to colibactin, also recapitulates the bacterial phenotype. In the course of this work, we discovered a novel cyclization pathway that was previously undetected in MS-based studies of colibactin, suggesting a refinement to the natural product structure and its mode of DNA binding. Colibactin 742 (4) and its precursor 9 will allow researchers to study colibactin's genotoxic effects independent of the producing organism for the first time.


Asunto(s)
Proteínas de Escherichia coli/síntesis química , Péptidos/síntesis química , Policétidos/síntesis química , ADN/química , Escherichia coli/genética , Humanos , Microbiota/genética , Conformación Molecular , Familia de Multigenes , Mutágenos/metabolismo , Mutación , Oxidación-Reducción , Fenotipo , Unión Proteica , Relación Estructura-Actividad
20.
ACS Chem Biol ; 16(7): 1243-1254, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34232632

RESUMEN

The complex reservoir of metabolite-producing bacteria in the gastrointestinal tract contributes tremendously to human health and disease. Bacterial composition, and by extension gut metabolomic composition, is undoubtably influenced by the use of modern antibiotics. Herein, we demonstrate that polymyxin B, a last resort antibiotic, influences the production of the genotoxic metabolite colibactin from adherent-invasive Escherichia coli (AIEC) NC101. Colibactin can promote colorectal cancer through DNA double stranded breaks and interstrand cross-links. While the structure and biosynthesis of colibactin have been elucidated, chemical-induced regulation of its biosynthetic gene cluster and subsequent production of the genotoxin by E. coli are largely unexplored. Using a multiomic approach, we identified that polymyxin B stress enhances the abundance of colibactin biosynthesis proteins (Clb's) in multiple pks+ E. coli strains, including pro-carcinogenic AIEC, NC101; the probiotic strain, Nissle 1917; and the antibiotic testing strain, ATCC 25922. Expression analysis via qPCR revealed that increased transcription of clb genes likely contributes to elevated Clb protein levels in NC101. Enhanced production of Clb's by NC101 under polymyxin stress matched an increased production of the colibactin prodrug motif, a proxy for the mature genotoxic metabolite. Furthermore, E. coli with a heightened tolerance for polymyxin induced greater mammalian DNA damage, assessed by quantification of γH2AX staining in cultured intestinal epithelial cells. This study establishes a key link between the polymyxin B stress response and colibactin production in pks+ E. coli. Ultimately, our findings will inform future studies investigating colibactin regulation and the ability of seemingly innocuous commensal microbes to induce host disease.


Asunto(s)
Farmacorresistencia Bacteriana/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Péptidos/efectos de los fármacos , Polimixinas/farmacología , Animales , Evolución Biológica , Línea Celular , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Genes Bacterianos/efectos de los fármacos , Familia de Multigenes/efectos de los fármacos , Mutágenos/metabolismo , Péptido Sintasas/genética , Péptidos/metabolismo , Sintasas Poliquetidas/genética , Policétidos/metabolismo , Ratas , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA