Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Microbiome ; 12(1): 86, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730492

ABSTRACT

BACKGROUND: Parasitic helminths influence the composition of the gut microbiome. However, the microbiomes of individuals living in helminth-endemic regions are understudied. The Orang Asli, an indigenous population in Malaysia with high burdens of the helminth Trichuris trichiura, display microbiotas enriched in Clostridiales, an order of spore-forming obligate anaerobes with immunogenic properties. We previously isolated novel Clostridiales that were enriched in these individuals and found that a subset promoted the Trichuris life cycle. In this study, we aimed to further characterize the functional properties of these bacteria. RESULTS: Clostridiales isolates were profiled for their ability to perform 57 enzymatic reactions and produce short-chain fatty acids (SCFAs) and hydrogen sulfide, revealing that these bacteria were capable of a range of activities associated with metabolism and host response. Consistent with this finding, monocolonization of mice with individual isolates identified bacteria that were potent inducers of regulatory T-cell (Treg) differentiation in the colon. Comparisons between variables revealed by these studies identified enzymatic properties correlated with Treg induction and Trichuris egg hatching. CONCLUSION: We identified Clostridiales species that are sufficient to induce high levels of Tregs. We also identified a set of metabolic activities linked with Treg differentiation and Trichuris egg hatching mediated by these newly isolated bacteria. Altogether, this study provides functional insights into the microbiotas of individuals residing in a helminth-endemic region. Video Abstract.


Subject(s)
Cell Differentiation , Clostridiales , Gastrointestinal Microbiome , T-Lymphocytes, Regulatory , Trichuris , Animals , T-Lymphocytes, Regulatory/immunology , Mice , Malaysia , Clostridiales/isolation & purification , Humans , Fatty Acids, Volatile/metabolism , Female , Trichuriasis/parasitology , Trichuriasis/immunology , Trichuriasis/microbiology
2.
Cell Host Microbe ; 32(5): 661-675.e10, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38657606

ABSTRACT

The intestine and liver are thought to metabolize dietary nutrients and regulate host nutrient homeostasis. Here, we find that the gut microbiota also reshapes the host amino acid (aa) landscape via efficiently metabolizing intestinal aa. To identify the responsible microbes/genes, we developed a metabolomics-based assay to screen 104 commensals and identified candidates that efficiently utilize aa. Using genetics, we identified multiple responsible metabolic genes in phylogenetically diverse microbes. By colonizing germ-free mice with the wild-type strain and their isogenic mutant deficient in individual aa-metabolizing genes, we found that these genes regulate the availability of gut and circulatory aa. Notably, microbiota genes for branched-chain amino acids (BCAAs) and tryptophan metabolism indirectly affect host glucose homeostasis via peripheral serotonin. Collectively, at single-gene level, this work characterizes a microbiota-encoded metabolic activity that affects host nutrient homeostasis and provides a roadmap to interrogate microbiota-dependent activity to improve human health.


Subject(s)
Amino Acids, Branched-Chain , Amino Acids , Gastrointestinal Microbiome , Homeostasis , Tryptophan , Animals , Gastrointestinal Microbiome/physiology , Mice , Amino Acids/metabolism , Amino Acids, Branched-Chain/metabolism , Tryptophan/metabolism , Mice, Inbred C57BL , Nutrients/metabolism , Intestines/microbiology , Humans , Metabolomics , Glucose/metabolism , Serotonin/metabolism , Germ-Free Life , Bacteria/metabolism , Bacteria/genetics , Bacteria/classification , Male
3.
J Exp Med ; 221(5)2024 May 06.
Article in English | MEDLINE | ID: mdl-38506708

ABSTRACT

Innate lymphoid cells (ILCs) can promote host defense, chronic inflammation, or tissue protection and are regulated by cytokines and neuropeptides. However, their regulation by diet and microbiota-derived signals remains unclear. We show that an inulin fiber diet promotes Tph1-expressing inflammatory ILC2s (ILC2INFLAM) in the colon, which produce IL-5 but not tissue-protective amphiregulin (AREG), resulting in the accumulation of eosinophils. This exacerbates inflammation in a murine model of intestinal damage and inflammation in an ILC2- and eosinophil-dependent manner. Mechanistically, the inulin fiber diet elevated microbiota-derived bile acids, including cholic acid (CA) that induced expression of ILC2-activating IL-33. In IBD patients, bile acids, their receptor farnesoid X receptor (FXR), IL-33, and eosinophils were all upregulated compared with controls, implicating this diet-microbiota-ILC2 axis in human IBD pathogenesis. Together, these data reveal that dietary fiber-induced changes in microbial metabolites operate as a rheostat that governs protective versus pathologic ILC2 responses with relevance to precision nutrition for inflammatory diseases.


Subject(s)
Immunity, Innate , Inflammatory Bowel Diseases , Humans , Animals , Mice , Interleukin-33 , Inulin , Lymphocytes , Dietary Fiber , Bile Acids and Salts , Inflammation
4.
Cell Rep Med ; 5(3): 101431, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38378002

ABSTRACT

Sulfasalazine is a prodrug known to be effective for the treatment of inflammatory bowel disease (IBD)-associated peripheral spondyloarthritis (pSpA), but the mechanistic role for the gut microbiome in regulating its clinical efficacy is not well understood. Here, treatment of 22 IBD-pSpA subjects with sulfasalazine identifies clinical responders with a gut microbiome enriched in Faecalibacterium prausnitzii and the capacity for butyrate production. Sulfapyridine promotes butyrate production and transcription of the butyrate synthesis gene but in F. prausnitzii in vitro, which is suppressed by excess folate. Sulfasalazine therapy enhances fecal butyrate production and limits colitis in wild-type and gnotobiotic mice colonized with responder, but not non-responder, microbiomes. F. prausnitzii is sufficient to restore sulfasalazine protection from colitis in gnotobiotic mice colonized with non-responder microbiomes. These findings reveal a mechanistic link between the efficacy of sulfasalazine therapy and the gut microbiome with the potential to guide diagnostic and therapeutic approaches for IBD-pSpA.


Subject(s)
Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Mice , Animals , Sulfasalazine/pharmacology , Sulfasalazine/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Treatment Outcome , Butyrates
5.
Front Physiol ; 14: 1290820, 2023.
Article in English | MEDLINE | ID: mdl-37954444

ABSTRACT

Colorectal cancer is a common malignant tumor in the gastrointestinal tract, which usually evolves from adenomatous polyps. However, due to the similarity in color between polyps and their surrounding tissues in colonoscopy images, and their diversity in size, shape, and texture, intelligent diagnosis still remains great challenges. For this reason, we present a novel dense residual-inception network (DRI-Net) which utilizes U-Net as the backbone. Firstly, in order to increase the width of the network, a modified residual-inception block is designed to replace the traditional convolutional, thereby improving its capacity and expressiveness. Moreover, the dense connection scheme is adopted to increase the network depth so that more complex feature inputs can be fitted. Finally, an improved down-sampling module is built to reduce the loss of image feature information. For fair comparison, we validated all method on the Kvasir-SEG dataset using three popular evaluation metrics. Experimental results consistently illustrates that the values of DRI-Net on IoU, Mcc and Dice attain 77.72%, 85.94% and 86.51%, which were 1.41%, 0.66% and 0.75% higher than the suboptimal model. Similarly, through ablation studies, it also demonstrated the effectiveness of our approach in colorectal semantic segmentation.

6.
Trends Microbiol ; 31(11): 1143-1161, 2023 11.
Article in English | MEDLINE | ID: mdl-37394299

ABSTRACT

Studies of the gut microbiota have revealed associations between specific bacterial species or community compositions with health and disease, yet the causal mechanisms underlying microbiota gene-host interactions remain poorly understood. This is partly due to limited genetic manipulation (GM) tools for gut bacteria. Here, we review current advances and challenges in the development of GM approaches, including clustered regularly interspaced short palindromic repeats (CRISPR)-Cas and transposase-based systems in either model or non-model gut bacteria. By overcoming barriers to 'taming' the gut microbiome, GM tools allow molecular understanding of host-microbiome associations and accelerate microbiome engineering for clinical treatment of cancer and metabolic disorders. Finally, we provide perspectives on the future development of GM for gut microbiome species, where more effort should be placed on assembling a generalized GM pipeline to accelerate the application of groundbreaking GM tools in non-model gut bacteria towards both basic understanding and clinical translation.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Bacteria/genetics , Gastrointestinal Microbiome/genetics , Host Microbial Interactions , CRISPR-Cas Systems/genetics
7.
bioRxiv ; 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37333296

ABSTRACT

Parasitic helminths influence the composition of the gut microbiome. However, the microbiomes of individuals living in helminth-endemic regions are understudied. The Orang Asli, an indigenous population in Malaysia with high burdens of the helminth Trichuris trichiura, displayed microbiotas enriched in Clostridiales, an order of spore-forming obligate anaerobes previously shown to have immunogenic properties. We previously isolated novel Clostridiales that were enriched in these individuals and found that a subset promoted the Trichuris life cycle. Here, we further characterized the functional properties of these bacteria. Enzymatic and metabolomic profiling revealed a range of activities associated with metabolism and host response. Consistent with this finding, monocolonization of mice with individual isolates identified bacteria that were potent inducers of regulatory T cell (Treg) differentiation in the colon. Comparisons between variables revealed by these studies identified enzymatic properties correlated with Treg induction and Trichuris egg hatching. These results provide functional insights into the microbiotas of an understudied population.

8.
Nature ; 611(7936): 578-584, 2022 11.
Article in English | MEDLINE | ID: mdl-36323778

ABSTRACT

Dietary fibres can exert beneficial anti-inflammatory effects through microbially fermented short-chain fatty acid metabolites<sup>1,2</sup>, although the immunoregulatory roles of most fibre diets and their microbiota-derived metabolites remain poorly defined. Here, using microbial sequencing and untargeted metabolomics, we show that a diet of inulin fibre alters the composition of the mouse microbiota and the levels of microbiota-derived metabolites, notably bile acids. This metabolomic shift is associated with type 2 inflammation in the intestine and lungs, characterized by IL-33 production, activation of group 2 innate lymphoid cells and eosinophilia. Delivery of cholic acid mimics inulin-induced type 2 inflammation, whereas deletion of the bile acid receptor farnesoid X receptor diminishes the effects of inulin. The effects of inulin are microbiota dependent and were reproduced in mice colonized with human-derived microbiota. Furthermore, genetic deletion of a bile-acid-metabolizing enzyme in one bacterial species abolishes the ability of inulin to trigger type 2 inflammation. Finally, we demonstrate that inulin enhances allergen- and helminth-induced type 2 inflammation. Taken together, these data reveal that dietary inulin fibre triggers microbiota-derived cholic acid and type 2 inflammation at barrier surfaces with implications for understanding the pathophysiology of allergic inflammation, tissue protection and host defence.


Subject(s)
Bile Acids and Salts , Dietary Fiber , Gastrointestinal Microbiome , Inflammation , Inulin , Animals , Humans , Mice , Bile Acids and Salts/metabolism , Cholic Acid/pharmacology , Dietary Fiber/pharmacology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Immunity, Innate , Inflammation/chemically induced , Inflammation/classification , Inflammation/pathology , Inulin/pharmacology , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/immunology , Metabolomics , Lung/drug effects , Lung/pathology , Intestines/drug effects , Intestines/microbiology , Intestines/pathology , Interleukin-33/metabolism , Eosinophils/cytology , Eosinophils/drug effects , Eosinophils/immunology
9.
Cell ; 185(22): 4170-4189.e20, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36240781

ABSTRACT

Nociceptive pain is a hallmark of many chronic inflammatory conditions including inflammatory bowel diseases (IBDs); however, whether pain-sensing neurons influence intestinal inflammation remains poorly defined. Employing chemogenetic silencing, adenoviral-mediated colon-specific silencing, and pharmacological ablation of TRPV1+ nociceptors, we observed more severe inflammation and defective tissue-protective reparative processes in a murine model of intestinal damage and inflammation. Disrupted nociception led to significant alterations in the intestinal microbiota and a transmissible dysbiosis, while mono-colonization of germ-free mice with Gram+Clostridium spp. promoted intestinal tissue protection through a nociceptor-dependent pathway. Mechanistically, disruption of nociception resulted in decreased levels of substance P, and therapeutic delivery of substance P promoted tissue-protective effects exerted by TRPV1+ nociceptors in a microbiota-dependent manner. Finally, dysregulated nociceptor gene expression was observed in intestinal biopsies from IBD patients. Collectively, these findings indicate an evolutionarily conserved functional link between nociception, the intestinal microbiota, and the restoration of intestinal homeostasis.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Mice , Animals , Gastrointestinal Microbiome/physiology , Nociceptors/physiology , Substance P , Dysbiosis , Inflammation
10.
Nat Commun ; 13(1): 6239, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36266345

ABSTRACT

The systemic metabolic shifts that occur during aging and the local metabolic alterations of a tumor, its stroma and their communication cooperate to establish a unique tumor microenvironment (TME) fostering cancer progression. Here, we show that methylmalonic acid (MMA), an aging-increased oncometabolite also produced by aggressive cancer cells, activates fibroblasts in the TME, which reciprocally secrete IL-6 loaded extracellular vesicles (EVs) that drive cancer progression, drug resistance and metastasis. The cancer-associated fibroblast (CAF)-released EV cargo is modified as a result of reactive oxygen species (ROS) generation and activation of the canonical and noncanonical TGFß signaling pathways. EV-associated IL-6 functions as a stroma-tumor messenger, activating the JAK/STAT3 and TGFß signaling pathways in tumor cells and promoting pro-aggressive behaviors. Our findings define the role of MMA in CAF activation to drive metastatic reprogramming, unveiling potential therapeutic avenues to target MMA at the nexus of aging, the tumor microenvironment and metastasis.


Subject(s)
Cancer-Associated Fibroblasts , Extracellular Vesicles , Neoplasms , Humans , Cancer-Associated Fibroblasts/metabolism , Reactive Oxygen Species/metabolism , Methylmalonic Acid/metabolism , Interleukin-6/metabolism , Tumor Microenvironment , Neoplasms/pathology , Extracellular Vesicles/metabolism , Transforming Growth Factor beta/metabolism
11.
Cell ; 185(3): 547-562.e22, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35051369

ABSTRACT

Hundreds of microbiota genes are associated with host biology/disease. Unraveling the causal contribution of a microbiota gene to host biology remains difficult because many are encoded by nonmodel gut commensals and not genetically targetable. A general approach to identify their gene transfer methodology and build their gene manipulation tools would enable mechanistic dissections of their impact on host physiology. We developed a pipeline that identifies the gene transfer methods for multiple nonmodel microbes spanning five phyla, and we demonstrated the utility of their genetic tools by modulating microbiome-derived short-chain fatty acids and bile acids in vitro and in the host. In a proof-of-principle study, by deleting a commensal gene for bile acid synthesis in a complex microbiome, we discovered an intriguing role of this gene in regulating colon inflammation. This technology will enable genetically engineering the nonmodel gut microbiome and facilitate mechanistic dissection of microbiota-host interactions.


Subject(s)
Gastrointestinal Microbiome/genetics , Genes, Bacterial , Animals , Bile Acids and Salts/metabolism , CRISPR-Cas Systems/genetics , Clostridium/genetics , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Dextran Sulfate , Drug Resistance, Microbial/genetics , Female , Gene Expression Regulation, Bacterial , Gene Transfer Techniques , Germ-Free Life , Inflammation/pathology , Intestines/pathology , Male , Metabolome/genetics , Metagenomics , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Insertional/genetics , Mutation/genetics , RNA, Ribosomal, 16S/genetics , Transcription, Genetic
12.
Gastroenterology ; 162(1): 166-178, 2022 01.
Article in English | MEDLINE | ID: mdl-34606847

ABSTRACT

BACKGROUND & AIMS: Fecal microbiota transplantation (FMT) is an emerging treatment modality for ulcerative colitis (UC). Several randomized controlled trials have shown efficacy for FMT in the treatment of UC, but a better understanding of the transferable microbiota and their immune impact is needed to develop more efficient microbiome-based therapies for UC. METHODS: Metagenomic analysis and strain tracking was performed on 60 donor and recipient samples receiving FMT for active UC. Sorting and sequencing of immunoglobulin (Ig) A-coated microbiota (called IgA-seq) was used to define immune-reactive microbiota. Colonization of germ-free or genetically engineered mice with patient-derived strains was performed to determine the mechanism of microbial impact on intestinal immunity. RESULTS: Metagenomic analysis defined a core set of donor-derived transferable bacterial strains in UC subjects achieving clinical response, which predicted response in an independent trial of FMT for UC. IgA-seq of FMT recipient samples and gnotobiotic mice colonized with donor microbiota identified Odoribacter splanchnicus as a transferable strain shaping mucosal immunity, which correlated with clinical response and the induction of mucosal regulatory T cells. Colonization of mice with O splanchnicus led to an increase in Foxp3+/RORγt+ regulatory T cells, induction of interleukin (IL) 10, and production of short chain fatty acids, all of which were required for O splanchnicus to limit colitis in mouse models. CONCLUSIONS: This work provides the first evidence of transferable, donor-derived strains that correlate with clinical response to FMT in UC and reveals O splanchnicus as a key component promoting both metabolic and immune cell protection from colitis. These mechanistic features will help enable strategies to enhance the efficacy of microbial therapy for UC. Clinicaltrials.gov ID NCT02516384.


Subject(s)
Bacteroidetes/immunology , Colitis/therapy , Colon/microbiology , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Immunoglobulin A/immunology , Intestinal Mucosa/microbiology , Animals , Bacteroidetes/genetics , Bacteroidetes/metabolism , Clinical Trials as Topic , Colitis/immunology , Colitis/metabolism , Colitis/microbiology , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/immunology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Colon/immunology , Colon/metabolism , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/immunology , Germ-Free Life , Humans , Immunity, Mucosal , Immunoglobulin A/genetics , Immunoglobulin A/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Intraepithelial Lymphocytes/microbiology , Metagenome , Metagenomics , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/microbiology , Treatment Outcome
13.
Cell Host Microbe ; 29(4): 607-619.e8, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33539767

ABSTRACT

Adherent-invasive E. coli (AIEC) are enriched in the intestinal microbiota of patients with Crohn's disease (CD) and promote intestinal inflammation. Yet, how AIEC metabolism of nutrients impacts intestinal homeostasis is poorly defined. Here, we show that AIEC encoding the large subunit of propanediol dehydratase (PduC), which facilitates the utilization of fucose fermentation product 1,2-propanediol, are increased in the microbiome of CD patients and drive AIEC-induced intestinal T cell inflammation. In murine models, CX3CR1+ mononuclear phagocytes (MNP) are required for PduC-dependent induction of T helper 17 (Th17) cells and interleukin-1ß (IL-1ß) production that leads to AIEC-induced inflammatory colitis. Activation of this inflammatory cascade requires the catalytic activity of PduC to generate propionate, which synergizes with lipopolysaccharide (LPS) to induce IL-1ß by MNPs. Disrupting fucose availability limits AIEC-induced propionate production and intestinal inflammation. These findings identify MNPs as metabolic sensors linking AIEC metabolism with intestinal inflammation and identify microbial metabolism as a potential therapeutic target in Crohn's disease treatment.


Subject(s)
Crohn Disease/metabolism , Escherichia coli Infections/metabolism , Escherichia coli/metabolism , Inflammation/metabolism , Intestines/immunology , Phagocytes/metabolism , Propylene Glycols/metabolism , Animals , Bacterial Adhesion , Crohn Disease/microbiology , Escherichia coli Infections/microbiology , Female , Host-Pathogen Interactions , Humans , Immunity , Interleukin-1beta , Intestinal Mucosa/metabolism , Intestines/microbiology , Male , Mice , Phagocytes/immunology , Th17 Cells
14.
Nature ; 581(7809): 475-479, 2020 05.
Article in English | MEDLINE | ID: mdl-32461639

ABSTRACT

Intestinal health relies on the immunosuppressive activity of CD4+ regulatory T (Treg) cells1. Expression of the transcription factor Foxp3 defines this lineage, and can be induced extrathymically by dietary or commensal-derived antigens in a process assisted by a Foxp3 enhancer known as conserved non-coding sequence 1 (CNS1)2-4. Products of microbial fermentation including butyrate facilitate the generation of peripherally induced Treg (pTreg) cells5-7, indicating that metabolites shape the composition of the colonic immune cell population. In addition to dietary components, bacteria modify host-derived molecules, generating a number of biologically active substances. This is epitomized by the bacterial transformation of bile acids, which creates a complex pool of steroids8 with a range of physiological functions9. Here we screened the major species of deconjugated bile acids for their ability to potentiate the differentiation of pTreg cells. We found that the secondary bile acid 3ß-hydroxydeoxycholic acid (isoDCA) increased Foxp3 induction by acting on dendritic cells (DCs) to diminish their immunostimulatory properties. Ablating one receptor, the farnesoid X receptor, in DCs enhanced the generation of Treg cells and imposed a transcriptional profile similar to that induced by isoDCA, suggesting an interaction between this bile acid and nuclear receptor. To investigate isoDCA in vivo, we took a synthetic biology approach and designed minimal microbial consortia containing engineered Bacteroides strains. IsoDCA-producing consortia increased the number of colonic RORγt-expressing Treg cells in a CNS1-dependent manner, suggesting enhanced extrathymic differentiation.


Subject(s)
Bacteria/metabolism , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Amino Acid Sequence , Animals , Bacteroides/metabolism , Colon/microbiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Fermentation , Gastrointestinal Microbiome , Male , Mice , Mice, Inbred C57BL , Microbial Consortia , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism
16.
Nat Prod Rep ; 37(1): 17-28, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31290896

ABSTRACT

Covering: 2012 to 2019HemN-like radical S-adenosyl-l-methionine (SAM) enzymes have been recently disclosed to catalyze diverse chemically challenging reactions from primary to secondary metabolic pathways. In this highlight, we summarize the reaction examples catalyzed by HemN-like enzymes to date and the enzymatic mechanisms reported. From the recent mechanistic investigations, we reason that there is a shared initiating mechanism wherein a characteristic SAM methylene radical is proposed to abstract a hydrogen atom from an sp3 carbon or add onto an sp2 carbon center although variations occur thereafter from reaction to reaction, as well as providing a brief insight into some future prospects.


Subject(s)
Enzymes/chemistry , Enzymes/metabolism , S-Adenosylmethionine/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Coproporphyrinogen Oxidase/chemistry , Coproporphyrinogen Oxidase/metabolism , Duocarmycins/metabolism , Escherichia coli Proteins/metabolism , Heme/metabolism , Hydrogen , Methylation , Peptides, Cyclic/metabolism , Polyketides/metabolism , Protein Methyltransferases/metabolism , Thiazoles/metabolism
17.
Org Lett ; 21(5): 1374-1378, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30763106

ABSTRACT

The biosynthetic gene clusters for herbicidins ( hbc) and aureonuclemycin ( anm) were identified in Streptomyces sp. KIB-027 and Streptomyces aureus, respectively. The roles of genes possibly involved in post-core-assembly steps in herbicidin biosynthesis in these clusters and a related her cluster were studied. Through systematic gene deletions, structural elucidation of the accumulated intermediates in the mutants, and in vitro verification of the encoded enzymes, the peripheral modification pathway for herbicidin biosynthesis is now fully established.


Subject(s)
Anti-Bacterial Agents/chemistry , Purine Nucleosides/biosynthesis , Streptomyces/chemistry , Molecular Structure
18.
Nat Commun ; 9(1): 2771, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30018376

ABSTRACT

Cyclopropanation of unactivated olefinic bonds via addition of a reactive one-carbon species is well developed in synthetic chemistry, whereas natural cyclopropane biosynthesis employing this strategy is very limited. Here, we identify a two-component cyclopropanase system, composed of a HemN-like radical S-adenosyl-L-methionine (SAM) enzyme C10P and a methyltransferase C10Q, catalyzes chemically challenging cyclopropanation in the antitumor antibiotic CC-1065 biosynthesis. C10P uses its [4Fe-4S] cluster for reductive cleavage of the first SAM to yield a highly reactive 5'-deoxyadenosyl radical, which abstracts a hydrogen from the second SAM to produce a SAM methylene radical that adds to an sp2-hybridized carbon of substrate to form a SAM-substrate adduct. C10Q converts this adduct to CC-1065 via an intramolecular SN2 cyclization mechanism with elimination of S-adenosylhomocysteine. This cyclopropanation strategy not only expands the enzymatic reactions catalyzed by the radical SAM enzymes and methyltransferases, but also sheds light on previously unnoticed aspects of the versatile SAM-based biochemistry.


Subject(s)
Antibiotics, Antineoplastic/biosynthesis , Bacterial Proteins/metabolism , Cyclopropanes/metabolism , Indoles/metabolism , Methyltransferases/metabolism , S-Adenosylmethionine/metabolism , Streptomyces/enzymology , Bacterial Proteins/genetics , Biocatalysis , Cloning, Molecular , Duocarmycins , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Iron/metabolism , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Methyltransferases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S-Adenosylhomocysteine/metabolism , Streptomyces/genetics , Sulfur/metabolism
20.
Nat Commun ; 8(1): 1485, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29133784

ABSTRACT

GyrI-like proteins are widely distributed in prokaryotes and eukaryotes, and recognized as small-molecule binding proteins. Here, we identify a subfamily of these proteins as cyclopropanoid cyclopropyl hydrolases (CCHs) that can catalyze the hydrolysis of the potent DNA-alkylating agents yatakemycin (YTM) and CC-1065. Co-crystallography and molecular dynamics simulation analyses reveal that these CCHs share a conserved aromatic cage for the hydrolytic activity. Subsequent cytotoxic assays confirm that CCHs are able to protect cells against YTM. Therefore, our findings suggest that the evolutionarily conserved GyrI-like proteins confer cellular protection against diverse xenobiotics via not only binding, but also catalysis.


Subject(s)
Alkylating Agents/chemistry , Bacterial Physiological Phenomena , Biocatalysis , Drug Resistance, Bacterial/physiology , Hydrolases/metabolism , Alkylating Agents/pharmacology , Animals , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , DNA/metabolism , DNA Topoisomerases, Type II/metabolism , Duocarmycins , Escherichia coli/physiology , Escherichia coli Proteins/metabolism , Humans , Hydrolases/chemistry , Hydrolases/genetics , Hydrolysis , Indoles/chemistry , Indoles/pharmacology , Inhibitory Concentration 50 , Jurkat Cells , Mice , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Pyrroles/chemistry , Pyrroles/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Sequence Homology, Amino Acid , Streptomyces/physiology , Xenobiotics/chemistry , Xenobiotics/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...