Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.792
1.
Sci Rep ; 14(1): 9432, 2024 04 24.
Article En | MEDLINE | ID: mdl-38658766

Novel psychoactive substances (NPS) represent a broad class of drugs new to the illicit market that often allow passing drug-screening tests. They are characterized by a variety of structures, rapid transience on the drug scene and mostly unknown metabolic profiles, thus creating an ever-changing scenario with evolving analytical targets. The present study aims at developing an indirect screening strategy for NPS monitoring, and specifically for new synthetic opioids (NSOs), based on assessing changes in endogenous urinary metabolite levels as a consequence of the systemic response following their intake. The experimental design involved in-vivo mice models: 16 animals of both sex received a single administration of morphine or fentanyl. Urine was collected before and after administration at different time points; the samples were then analysed with an untargeted metabolomics LC-HRMS workflow. According to our results, the intake of opioids resulted in an elevated energy demand, that was more pronounced on male animals, as evidenced by the increase in medium and long chain acylcarnitines levels. It was also shown that opioid administration disrupted the pathways related to catecholamines biosynthesis. The observed alterations were common to both morphine and fentanyl: this evidence indicate that they are not related to the chemical structure of the drug, but rather on the drug class. The proposed strategy may reinforce existing NPS screening approaches, by identifying indirect markers of drug assumption.


Analgesics, Opioid , Fentanyl , Metabolomics , Morphine , Animals , Male , Female , Mice , Metabolomics/methods , Analgesics, Opioid/urine , Fentanyl/analogs & derivatives , Fentanyl/urine , Fentanyl/metabolism , Chromatography, High Pressure Liquid/methods , Morphine/urine , Psychotropic Drugs/urine , Mass Spectrometry/methods , Metabolome/drug effects
2.
Cells ; 13(8)2024 Apr 09.
Article En | MEDLINE | ID: mdl-38667276

Cancer cell lines are frequently used in metabolomics, such as in vitro tumor models. In particular, A2780 cells are commonly used as a model for ovarian cancer to evaluate the effects of drug treatment. Here, we compare the NMR metabolomics profiles of A2780 and cisplatin-resistant A2780 cells with those of cells derived from 10 patients with high-grade serous ovarian carcinoma (collected during primary cytoreduction before any chemotherapeutic treatment). Our analysis reveals a substantial similarity among all primary cells but significant differences between them and both A2780 and cisplatin-resistant A2780 cells. Notably, the patient-derived cells are closer to the resistant A2780 cells when considering the exo-metabolome, whereas they are essentially equidistant from A2780 and A2780-resistant cells in terms of the endo-metabolome. This behavior results from dissimilarities in the levels of several metabolites attributable to the differential modulation of underlying biochemical pathways. The patient-derived cells are those with the most pronounced glycolytic phenotype, whereas A2780-resistant cells mainly diverge from the others due to alterations in a few specific metabolites already known as markers of resistance.


Cisplatin , Drug Resistance, Neoplasm , Magnetic Resonance Spectroscopy , Metabolomics , Ovarian Neoplasms , Humans , Female , Cisplatin/pharmacology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Metabolomics/methods , Cell Line, Tumor , Magnetic Resonance Spectroscopy/methods , Metabolome/drug effects , Antineoplastic Agents/pharmacology
3.
J Microbiol Biotechnol ; 34(4): 828-837, 2024 Apr 28.
Article En | MEDLINE | ID: mdl-38668685

Vancomycin (VAN) and metronidazole (MTR) remain the current drugs of choice for the treatment of non-severe Clostridioides difficile infection (CDI); however, while their co-administration has appeared in clinical treatment, the efficacy varies greatly and the mechanism is unknown. In this study, a CDI mouse model was constructed to evaluate the therapeutic effects of VAN and MTR alone or in combination. For a perspective on the intestinal ecology, 16S rRNA amplicon sequencing and non-targeted metabolomics techniques were used to investigate changes in the fecal microbiota and metabolome of mice under the co-administration treatment. As a result, the survival rate of mice under co-administration was not dramatically different compared to that of single antibiotics, and the former caused intestinal tissue hyperplasia and edema. Co-administration also significantly enhanced the activity of amino acid metabolic pathways represented by phenylalanine, arginine, proline, and histidine, decreased the level of deoxycholic acid (DCA), and downregulated the abundance of beneficial microbes, such as Bifidobacterium and Akkermansia. VAN plays a dominant role in microbiota regulation in co-administration. In addition, co-administration reduced or increased the relative abundance of antibiotic-sensitive bacteria, including beneficial and harmful microbes, without a difference. Taken together, there are some risks associated with the co-administration of VAN and MTR, and this combination mode should be used with caution in CDI treatment.


Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Disease Models, Animal , Drug Therapy, Combination , Feces , Gastrointestinal Microbiome , Metronidazole , RNA, Ribosomal, 16S , Vancomycin , Animals , Metronidazole/administration & dosage , Vancomycin/administration & dosage , Vancomycin/pharmacology , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Gastrointestinal Microbiome/drug effects , Mice , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , RNA, Ribosomal, 16S/genetics , Feces/microbiology , Intestines/microbiology , Intestines/drug effects , Male , Bacteria/classification , Bacteria/genetics , Bacteria/drug effects , Metabolome/drug effects
4.
Sci Total Environ ; 927: 171851, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38518822

Untargeted metabolomics is a non-a priori analysis of biomolecules that characterizes the metabolome variations induced by short- and long-term exposures to stressors. Even if the metabolite annotation remains lacunar due to database gaps, the global metabolomic fingerprint allows for trend analyses of dose-response curves for hundreds of cellular metabolites. Analysis of dose/time-response curve trends (biphasic or monotonic) of untargeted metabolomic features would thus allow the use of all the chemical signals obtained in order to determine stress levels (defense or damage) in organisms. To develop this approach in a context of time-dependent microbial community changes, mature river biofilms were exposed for 1 month to four cobalt (Co) concentrations (from background concentration to 1 × 10-6 M) in an open system of artificial streams. The meta-metabolomic response of biofilms was compared against a multitude of biological parameters (including bioaccumulation, biomass, chlorophyll a content, composition and structure of prokaryotic and eukaryotic communities) monitored at set exposure times (from 1 h to 28 d). Cobalt exposure induced extremely rapid responses of the meta-metabolome, with time range inducing defense responses (TRIDeR) of around 10 s, and time range inducing damage responses (TRIDaR) of several hours. Even in biofilms whose structure had been altered by Co bioaccumulation (reduced biomass, chlorophyll a contents and changes in the composition and diversity of prokaryotic and eukaryotic communities), concentration range inducing defense responses (CRIDeR) with similar initiation thresholds (1.41 ± 0.77 × 10-10 M Co2+ added in the exposure medium) were set up at the meta-metabolome level at every time point. In contrast, the concentration range inducing damage responses (CRIDaR) initiation thresholds increased by 10 times in long-term Co exposed biofilms. The present study demonstrates that defense and damage responses of biofilm meta-metabolome exposed to Co are rapidly and sustainably impacted, even within tolerant and resistant microbial communities.


Biofilms , Cobalt , Metabolome , Rivers , Water Pollutants, Chemical , Biofilms/drug effects , Cobalt/toxicity , Rivers/microbiology , Water Pollutants, Chemical/toxicity , Metabolome/drug effects , Metabolomics , Microbiota/drug effects
5.
J Hazard Mater ; 470: 134099, 2024 May 15.
Article En | MEDLINE | ID: mdl-38547754

The response of the meta-metabolome is rarely used to characterize the effects of contaminants on a whole community. Here, the meta-metabolomic fingerprints of biofilms were examined after 1, 3 and 7 days of exposure to five concentrations of cobalt (from background concentration to 1 × 10-5 M) in aquatic microcosms. The untargeted metabolomic data were processed using the DRomics tool to build dose-response models and to calculate benchmark-doses. This approach made it possible to use 100% of the chemical signal instead of being limited to the very few annotated metabolites (7%). These benchmark-doses were further aggregated into an empirical cumulative density function. A trend analysis of the untargeted meta-metabolomic feature dose-response curves after 7 days of exposure suggested the presence of a concentration range inducing defense responses between 1.7 × 10-9 and 2.7 × 10-6 M, and of a concentration range inducing damage responses from 2.7 × 10-6 M and above. This distinction was in good agreement with changes in the other biological parameters studied (biomass and chlorophyll content). This study demonstrated that the molecular defense and damage responses can be related to contaminant concentrations and represents a promising approach for environmental risk assessment of metals.


Biofilms , Cobalt , Dose-Response Relationship, Drug , Rivers , Water Pollutants, Chemical , Cobalt/toxicity , Biofilms/drug effects , Water Pollutants, Chemical/analysis , Water Pollutants, Chemical/toxicity , Rivers/chemistry , Rivers/microbiology , Metabolomics , Metabolome/drug effects
6.
Food Funct ; 15(8): 4354-4364, 2024 Apr 22.
Article En | MEDLINE | ID: mdl-38533683

Alzheimer's Disease (AD) is a fatal age-related neurodegenerative condition with a multifactorial etiology contributing to 70% of dementia globally. The search for a multi-target agent to hit different targets involved in the pathogenesis of AD is crucial. In the present study, the neuroprotective effects of four Morus extracts were assessed in LPS-induced AD in mice. Among the studied species, M. macroura exhibited a profound effect on alleviating the loss of cognitive function, improved the learning ability, restored the acetylcholine esterase (AChE) levels to normal, and significantly reduced the tumor necrosis factor alpha (TNF-α) brain content in LPS-treated mice. To investigate the secondary metabolome of the studied Morus species, ultra-performance liquid chromatography coupled to tandem mass spectrometry (UPLC-HRMS/MS), aided with feature-based molecular networking, was employed. Among the annotated features, aryl benzofurans and prenylated flavonoids were suggested as being responsible for the observed neuroprotective effect. Furthermore, some of the detected metabolites were proposed as new natural products such as moranoline di-O-hexoside (1), isomers of trimethoxy-dihydrochalcone-O-dihexoside (59 & 76), (hydroxy-dimethoxyphenyl)butenone-O-hexoside (82), and O-methylpreglabridin-O-sulphate (105). In conclusion, our findings advocate the potential usage of M. macroura leaves for the management of AD, yet after considering further clinical trials.


Alzheimer Disease , Metabolome , Morus , Neuroprotective Agents , Plant Extracts , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Neuroprotective Agents/pharmacology , Mice , Plant Extracts/pharmacology , Male , Morus/chemistry , Metabolome/drug effects , Tandem Mass Spectrometry , Disease Models, Animal , Chromatography, High Pressure Liquid , Humans , Brain/metabolism , Brain/drug effects
7.
J Appl Microbiol ; 135(4)2024 Apr 01.
Article En | MEDLINE | ID: mdl-38253409

AIMS: To examine the influence of GED on the gut microbiota and metabolites using a bilateral ovariectomized (OVX) rat model. We tried to elucidate the underlying mechanisms of GED in the treatment of menopausal hot flashes. METHODS AND RESULTS: 16S rRNA sequencing, metabonomics, molecular biological analysis, and fecal microbiota transplantation (FMT) were conducted to elucidate the mechanisms by which GED regulates the gut microbiota. GED significantly reduced OVX-induced hot flashes and improved disturbances in the gut microbiota metabolites. Moreover, FMT validated that the gut microbiota can trigger hot flashes, while GED can alleviate hot flash symptoms by modulating the composition of the gut microbiota. Specifically, GED upregulated the abundance of Blautia, thereby increasing l(+)-ornithine levels for the treatment of menopausal hot flashes. Additionally, GED affected endothelial nitric oxide synthase and heat shock protein 70 (HSP70) levels in the hypothalamic preoptic area by changing the gut microbiota composition. CONCLUSIONS: Our study illuminated the underlying mechanisms by which GED attenuated the hot flashes through modulation of the gut microbiota and explored the regulatory role of the gut microbiota on HSP70 expression in the preoptic anterior hypothalamus, thereby establishing a foundation for further exploration of the role of the gut-brain axis in hot flashes.


Drugs, Chinese Herbal , Gastrointestinal Microbiome , Hot Flashes , Menopause , Animals , Gastrointestinal Microbiome/drug effects , Hot Flashes/metabolism , Hot Flashes/drug therapy , Rats , Female , Drugs, Chinese Herbal/pharmacology , Fecal Microbiota Transplantation , Ovariectomy , Rats, Sprague-Dawley , RNA, Ribosomal, 16S/genetics , Metabolome/drug effects
8.
Schizophr Bull ; 49(3): 646-658, 2023 05 03.
Article En | MEDLINE | ID: mdl-36723169

BACKGROUND AND HYPOTHESIS: Antipsychotics (APs), the cornerstone of schizophrenia treatment, confer a relatively high risk of constipation. However, the mechanisms underpinning AP-induced constipation are poorly understood. Thus, we hypothesized that (1) schizophrenia patients with AP-induced constipation have distinct metabolic patterns; (2) there is more than one mechanism at play in producing this adverse drug effect; and (3) AP-associated changes in the gut microbiome are related to the altered metabolic profiles. STUDY DESIGN: Eighty-eight schizophrenia patients, including 44 with constipation (C) and 44 matched patients without constipation (NC), were enrolled in this study. Constipation was diagnosed by Rome IV criteria for constipation and colonic transit time using radiopaque markers (ROMs) while severity was evaluated with the Bristol Stool Form Scale (BSS) and Constipation Assessment Scale (CAS). Fasting blood samples were drawn from all participants and were subjected to non-targeted liquid chromatography-mass spectrometry (LC-MS) metabolomic analysis. STUDY RESULTS: Eleven metabolites were significantly altered in AP-induced constipation which primarily disturbed sphingolipid metabolism, choline metabolism, and sphingolipid signaling pathway (P value < .05, FDR < 0.05). In the C group, changes in the gut bacteria showed a certain degree of correlation with 2 of the significantly altered serum metabolites and were associated with alterations in choline metabolism. CONCLUSIONS: Our findings indicated that there were disturbances in distinct metabolic pathways that were associated with AP-induced constipation. In addition, this study presents evidence of a link between alterations in the gut microbiome and host metabolism which provides additional mechanistic insights on AP-induced constipation.


Antipsychotic Agents , Constipation , Schizophrenia , Humans , Antipsychotic Agents/adverse effects , Choline/metabolism , Constipation/chemically induced , Constipation/metabolism , Gastrointestinal Microbiome/drug effects , Metabolome/drug effects , Schizophrenia/blood , Schizophrenia/drug therapy , Schizophrenia/metabolism , Sphingolipids/metabolism , Case-Control Studies , Male , Female , Adult , Middle Aged
9.
Sci Rep ; 12(1): 11884, 2022 07 13.
Article En | MEDLINE | ID: mdl-35831335

Liver cirrhosis is a late-stage liver disease characterized by excessive fibrous deposition triggering portal-hypertension (PH); the prime restrainer for cirrhosis-related complications. Remedies that can dually oppose hepatic fibrosis and lower PH, may prevent progression into decompensated-cirrhosis. Different Astragalus-species members have shown antifibrotic and diuretic actions with possible subsequent PH reduction. However, A.spinosus and A.trigonus were poorly tested for eliciting these actions. Herein, A.spinosus and A.trigonus roots and aerial parts extracts were subjected to comprehensive metabolic-fingerprinting using UHPLC-MS/MS resulting in 56 identified phytoconstituents, followed by chemometric untargeted analysis that revealed variable metabolic profiles exemplified by different species and organ types. Consequently, tested extracts were in-vivo evaluated for potential antifibrotic/anticirrhotic activity by assessing specific markers. The mechanistic prospective to induce diuresis was investigated by analyzing plasma aldosterone and renal-transporters gene-expression. Serum apelin and dimethylarginine-dimethylaminohydrolase-1 were measured to indicate the overall effect on PH. All extracts amended cirrhosis and PH to varying extents and induced diuresis via different mechanisms. Further, An OPLS model was built to generate a comprehensive metabolic-profiling of A.spinosus and A.trigonus secondary-metabolites providing a chemical-based evidence for their efficacious consistency. In conclusion, A.spinosus and A.trigonus organs comprised myriad pharmacologically-active constituents that act synergistically to ameliorate cirrhosis and associated PH.


Astragalus Plant , Hypertension, Portal , Liver Cirrhosis , Plant Extracts , Aldosterone/blood , Amidohydrolases/blood , Apelin/blood , Astragalus Plant/chemistry , Astragalus Plant/metabolism , Chromatography, High Pressure Liquid , Diuresis , Hydrogen-Ion Concentration , Hypertension, Portal/blood , Hypertension, Portal/drug therapy , Hypertension, Portal/etiology , Hypertension, Portal/metabolism , Liver/metabolism , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Metabolome/drug effects , Phytochemicals/chemistry , Phytochemicals/metabolism , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Plant Extracts/chemistry , Plant Extracts/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Prospective Studies , Tandem Mass Spectrometry
10.
Article En | MEDLINE | ID: mdl-35245842

A new method involving gut microbiota biotransformation, spectrum-effect relationship analysis and metabolomics analysis was developed to study the antitussive and expectorant microbial metabolites of platycosides fraction (MPFs) of Platycodonis Radix. Furthermore, their possible metabolic mechanisms were studied for the first time. The findings showed that the antitussive and expectorant effects of the platycosides fraction (PF) were significantly enhanced by the gut microbiota biotransformation. 11 active antitussive microbial metabolites and 12 active expectorant microbial metabolites, which shared 8 components, were successfully screened out via spectrum-effect relationship analysis. The prototypes of the active microbial metabolites could be reversely traced according to the gut microbiota biotransformation pathways. It was found out that one platycoside could produce several active microbial metabolites and several different platycosides could produce the same active microbial metabolite. In addition, the metabolomics analysis showed that both the PF and its active microbial metabolites could regulate the same metabolomic pathways of Linoleic acid metabolism, Arachidonic acid metabolism and Glycerophospholipid metabolism to exert antitussive activity, and regulate the same metabolomic pathway of Arachidonic acid metabolism to exert expectorant activity. These findings suggested the microbial metabolites may be the active forms of the platycosides. Overall, the proposed approach was useful in screening the active microbial metabolites; this work explained the in vivo antitussive and expectorant metabolic mechanisms of multi-constituents, multi-targets and synergistic effects of PF of Platycodonis Radix.


Antitussive Agents , Expectorants , Metabolome/drug effects , Plant Extracts , Platycodon , Animals , Antitussive Agents/chemistry , Antitussive Agents/pharmacology , Chromatography, Liquid , Expectorants/chemistry , Expectorants/pharmacology , Gastrointestinal Microbiome , Metabolomics , Mice , Oleanolic Acid/analogs & derivatives , Plant Extracts/chemistry , Plant Extracts/pharmacology , Platycodon/chemistry , Saponins
11.
Cell ; 185(3): 513-529.e21, 2022 02 03.
Article En | MEDLINE | ID: mdl-35120663

The human gut microbiota resides within a diverse chemical environment challenging our ability to understand the forces shaping this ecosystem. Here, we reveal that fitness of the Bacteroidales, the dominant order of bacteria in the human gut, is an emergent property of glycans and one specific metabolite, butyrate. Distinct sugars serve as strain-variable fitness switches activating context-dependent inhibitory functions of butyrate. Differential fitness effects of butyrate within the Bacteroides are mediated by species-level variation in Acyl-CoA thioesterase activity and nucleotide polymorphisms regulating an Acyl-CoA transferase. Using in vivo multi-omic profiles, we demonstrate Bacteroides fitness in the human gut is associated together, but not independently, with Acyl-CoA transferase expression and butyrate. Our data reveal that each strain of the Bacteroides exists within a unique fitness landscape based on the interaction of chemical components unpredictable by the effect of each part alone mediated by flexibility in the core genome.


Gastrointestinal Microbiome , Metabolome , Polysaccharides/metabolism , Acyl Coenzyme A/metabolism , Amino Acid Sequence , Amino Acids, Branched-Chain/metabolism , Bacteroidetes/drug effects , Bacteroidetes/genetics , Bacteroidetes/growth & development , Butyrates/chemistry , Butyrates/pharmacology , Coenzyme A-Transferases/chemistry , Coenzyme A-Transferases/metabolism , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Genetic Variation/drug effects , Hydrogen-Ion Concentration , Metabolome/drug effects , Metabolome/genetics , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Species Specificity , Stress, Physiological/drug effects , Stress, Physiological/genetics , Transcription, Genetic/drug effects
12.
Sci Rep ; 12(1): 2066, 2022 02 08.
Article En | MEDLINE | ID: mdl-35136077

In this study, we utilized an untargeted NMR metabolomics approach to identify the vector response in terms of metabolic profiling after temperature and insecticide exposure in comparison with the control. Clearly, temperature and insecticide exposure cause changes in the underlying metabolism, and the NMR metabolomic profile enables a direct examination of the immediate response of the vector to cope up with these changes. The present study was designed in four parts: A-Aedes aegypti were exposed to 40 °C for one-hour, DDT-4%, malathion-5%, and deltamethrin-0.05% separately and, part B-D; one-hour exposure at 35 °C and 40 °C temperatures followed by one-hour exposure to insecticide. The resultant metabolite profiles were compared with the control. In response to temperature and insecticide exposure, several metabolites and altered pathways were identified. Citrate, maltose, lipids, Nicotinate, Choline, Pyruvate and ß-hydroxybutyrate were found as important components of major biological pathways such as tri-carboxylic acid cycle, branched amino acid degradation, glycolysis/gluconeogenesis, amino acid metabolism, lipid and carbohydrate metabolism, nucleotide PRPP pathway, and phospholipid metabolism. Furthermore, the results also suggest that the changes imposed by exposure to temperature and insecticides individually, are reversed with combined exposure, thus negating the impact of each other and posing a threat to the control of Aedes-borne diseases such as dengue, chikungunya, Zika and yellow fever.


Aedes/metabolism , Insecticides/pharmacology , Metabolic Networks and Pathways/drug effects , Metabolome/drug effects , Vector Borne Diseases/transmission , Aedes/drug effects , Animals , DDT/pharmacology , Insecticide Resistance/physiology , Malathion/pharmacology , Metabolomics/methods , Mosquito Vectors/drug effects , Nitriles/pharmacology , Pyrethrins/pharmacology , Temperature
13.
Oxid Med Cell Longev ; 2022: 4636618, 2022.
Article En | MEDLINE | ID: mdl-35126813

Inflammatory bowel diseases (IBDs) constitute a group of chronic intestinal conditions prominently featuring deranged metabolism. Effective pharmacological treatments for IBDs are lacking. Isosteviol sodium (STV-Na) exhibits anti-inflammatory activity and may offer therapeutic benefits in chronic colitis. However, the associated mechanism remains unclear. This study is aimed at exploring the therapeutic effects of STV-Na against chronic colitis in terms of metabolic reprogramming and macrophage polarization. Results show that STV-Na attenuated weight loss and colonic pathological damage and restored the hematological and biochemical parameters in chronic colitis mice models. STV-Na also restored intestinal permeability by increasing the goblet cell numbers, which was accompanied by lowered plasma lipopolysaccharide and diamine oxidase levels. Metabolomic analysis highlighted 102 candidate biomarkers and 5 vital pathways that may be crucial in the potential pharmacological mechanism of STV-Na in regulating intestinal inflammation and oxidative stress. These pathways were glycerophospholipid metabolism, phenylalanine metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, the pentose phosphate pathway, and phosphonate and phosphinate metabolism. Furthermore, STV-Na significantly decreased M1 macrophage polarization in the spleen and colon. The mRNA and protein levels of IL-1ß, TNF-α, and NF-κB/p65 in colonic tissue from the colitis mice were decreased after the STV-Na treatment. Overall, STV-Na could alleviate chronic colitis by suppressing oxidative stress and inflammation levels, reprogramming the metabolic profile, inhibiting macrophage polarization, and suppressing the NF-κB/p65 signaling pathway. STV-Na remains a promising candidate drug for treating IBDs.


Colitis/pathology , Diterpenes, Kaurane/pharmacology , Macrophage Activation/drug effects , Metabolome/drug effects , Signal Transduction/drug effects , Animals , Chronic Disease , Colitis/chemically induced , Colitis/drug therapy , Colon/drug effects , Colon/metabolism , Colon/pathology , Dextran Sulfate/toxicity , Diterpenes, Kaurane/therapeutic use , Glycerophospholipids/metabolism , Interleukin-1beta/blood , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/metabolism , Male , Metabolomics , Mice , Mice, Inbred C57BL , Pentose Phosphate Pathway , Phenylalanine/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
14.
Molecules ; 27(4)2022 Feb 14.
Article En | MEDLINE | ID: mdl-35209070

Discovering new and effective drugs for the treatment of Alzheimer's disease (AD) is a major clinical challenge. This study focuses on chemical modulation of the gut microbiome in an established murine AD model. We used the 16S rDNA sequencing technique to investigate the effect of xanthohumol (Xn) on the diversity of intestinal microflora in 2-month- and 6-month-old APP/PS1 mice, respectively. APP/PS1 and wild-type mice were treated by gavage with corn oil with or without Xn every other day for 90 days. Prior to and following treatment, animals were tested for spatial learning, cognitive and memory function. We found Xn reduced cognitive dysfunction in APP/PS1 mice and significantly regulated the composition and abundance of gut microbiota both in prevention experiments (with younger mice) and therapeutic experiments (with older mice). Differential microflora Gammaproteobacteria were significantly enriched in APP/PS1 mice treated with Xn. Nodosilineaceae and Rikenellaceae may be the specific microflora modulated by Xn. The penicillin and cephalosporin biosynthesis pathway and the atrazine degradation pathway may be the principal modulation pathways. Taken together, oral treatment with Xn may have a neuroprotective role by regulating the composition of intestinal microflora, a result that contributes to the scientific basis for a novel prophylactic and therapeutic approach to AD.


Biological Products/pharmacology , Flavonoids/pharmacology , Gastrointestinal Microbiome/drug effects , Metabolome/drug effects , Propiophenones/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Biodiversity , Biological Products/chemistry , Cognition/drug effects , Disease Models, Animal , Flavonoids/chemistry , Metagenome , Metagenomics/methods , Mice , Mice, Transgenic , Propiophenones/chemistry
15.
Article En | MEDLINE | ID: mdl-35151210

Gut microbiota is associated with tumor progress and host metabolic disorder, but whether gut microbiota regulation can affect cancer growth through interfering host metabolism maintains unknown yet. Here, we used combined antibiotics (ABX) to build an extremely altered gut microbiota ecosystem and study its influence on the xenograft MC38 tumor as well as the associations of the effects with host metabolisms. The MC38 tumor bearing mouse was treated with ABX (vancomycin, neomycin and imipenem-cilastatin) to build the extremely altered microbiota ecosystem, the gut microbiota diversity alteration was determined by 16S rRNA based gene sequencing. The effects of the altered microbiota on tumor were assessed by cell apoptosis and growth rate of the tumor. The potential metabolic biomarkers and involved metabolism pathways were screened out by UPLC-QTOF-MS/MS based untargeted metabolomics and KEGG analysis respectively. The correlations between key metabolites and microbiota were analyzed by Spearman correlation analysis. Compared with the un-treated mice, the tumor growth of ABX-treated mice was significantly suppressed, and the cell apoptosis was obviously promoted. The gut microbiota diversity was decreased significantly with the dominant bacteria phylum Bacteroidetes and Firmicutes replaced by Proteobacteria, which involved 14 significantly altered bacteria genera. Four potential targeted metabolism pathways, including sphingolipid, glycerophospholipid, arginine-proline and primary bile acid metabolism, were screened out, and the involved key metabolites such as ceramide, phosphatidylethanolamine, phosphatidylcholine, taurocholic acid and L-proline were correlated significantly with the altered bacteria genera. Through the integrated analysis of microbiome and metabolomics, it was revealed that gut microbiota regulation may inhibit the xenograft MC38 tumor growth potentially by interfering host lipid and amino acid metabolisms, such as sphingolipid, glycerophospholipid, primary bile acid and arginine-proline metabolisms in this case.


Amino Acids/metabolism , Gastrointestinal Microbiome/drug effects , Lipid Metabolism , Metabolome/drug effects , Neoplasms, Experimental/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Male , Metabolomics , Mice , Mice, Inbred C57BL , Tandem Mass Spectrometry
16.
Toxicology ; 467: 153095, 2022 02 15.
Article En | MEDLINE | ID: mdl-34999168

Mefentrifluconazole, a new type of chiral triazole fungicide, is widely applied to control a variety of fungal diseases in crops. However, the toxicological effects of mefentrifluconazole on aquatic organisms are unknown, especially at the enantiomer level. In the present study, zebrafish were selected as a typical model for mefentrifluconazole enantiomer exposure. Metabolomic and transcription analyses were performed with 0.01 and 0.10 mg/L mefentrifluconazole and its enantiomers (i.e., rac-mfz/(-)-mfz/(+)-mfz) at 28 days. The 1H nuclear magnetic resonance (NMR)-based metabolomics analysis showed that 9, 10 and 4 metabolites were changed significantly in the rac-mfz, (+)-mfz and (-)-mfz treatment groups compared with the control group, respectively. The differential metabolites were related to energy metabolism, lipid metabolism and amino acid metabolism. The qRT-PCR analysis revealed that the expression of lipid metabolism-, apoptosis- and CYP-related genes in the livers of female zebrafish in rac-mfz and (+)-mfz was 1.61-108.92 times and 2.37-551.34 times higher than that in (-)-mfz, respectively. The results above indicate that exposure to mefentrifluconazole induced enantioselective liver toxicity in zebrafish. Our study underlined the importance of distinguishing different enantiomers, which will contribute to environmental protection.


Energy Metabolism/drug effects , Fluconazole/analogs & derivatives , Fungicides, Industrial/toxicity , Liver/drug effects , Metabolome/drug effects , Transcriptome/drug effects , Zebrafish , Animals , Female , Fluconazole/chemistry , Fluconazole/toxicity , Fungicides, Industrial/chemistry , Gene Expression Profiling , Gene Expression Regulation , Lipid Metabolism/drug effects , Liver/metabolism , Liver/pathology , Male , Metabolomics , Molecular Structure , Proton Magnetic Resonance Spectroscopy , Sex Factors , Stereoisomerism , Structure-Activity Relationship , Zebrafish/genetics , Zebrafish/metabolism
17.
Food Funct ; 13(4): 1808-1821, 2022 Feb 21.
Article En | MEDLINE | ID: mdl-35084009

Tripeptide NCW identified in our previous study displayed a strong ACE inhibitory activity, but whether it has any antihypertensive effect in vivo remains unknown. Thus, in this study, we aimed to investigate the protective effects of tripeptide NCW in spontaneously hypertensive rats (SHRs) and to further figure out the serum metabolic profiling variations due to its oral administration via UPLC-Q-TOF-MS/MS-based metabolomics analysis to clarify the underlying hypotensive mechanism. After three weeks of oral administration, the tripeptide NCW-treated group (NCW/SHR group, 80 mg per kg BW per d) showed significantly reduced systolic and diastolic blood pressure by 48.08 ± 3.84 mmHg and 48.92 ± 5.77 mmHg, respectively. Additionally, a total of 25 blood pressure-related metabolites were identified as being significantly changed in SHRs given tripeptide NCW after three weeks. These 25 metabolites might be biomarkers that indicated that the tripeptide NCW exhibits antihypertensive activity via regulating bile acid metabolism, lipid metabolism, amino acid metabolism, purinergic signaling, pantothenate and CoA biosynthesis, and the citrate cycle. Collectively, tripeptide NCW has a protective effect on SHRs associated with serum metabolite abnormalities.


Antihypertensive Agents , Hypertension/metabolism , Metabolome/drug effects , Oligopeptides , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Biomarkers/metabolism , Blood Pressure/drug effects , Lipid Metabolism/drug effects , Male , Metabolomics , Oligopeptides/chemistry , Oligopeptides/pharmacology , Protective Agents/chemistry , Protective Agents/pharmacology , Rats , Rats, Inbred SHR
18.
Food Funct ; 13(4): 1860-1880, 2022 Feb 21.
Article En | MEDLINE | ID: mdl-35084415

Intestinal microbiota and metabonomics were integrated to investigate the efficiency of non-saponification or saponification astaxanthin (N-Asta or S-Asta) derived from Penaeus sinensis by-products on alleviating paracetamol (PCM)-induced oxidative stress. Pre-treatment with N-Asta or S-Asta for 14 days restored the cellular morphology of the intestine and increased glutathione (GSH) levels under PCM overdose in rats. However, S-Asta displayed higher adsorption than that of N-Asta. PCM overdose reduced the richness and diversity of intestinal microbiota in the model group. Comparably, N-Asta or S-Asta pre-treatment increased the Actinobacteria abundance. Increased phyla Bacteroidetes and Verrucomicrobia were only found in the S-Asta-pre-treated group. At the genus level, N-Asta pre-treatment increased Lactobacillus and Parasutterella abundance, whereas S-Asta pre-treatment elevated Bacteroidales_S24-7_group_norank and Ruminococcaceae_uncultured. Compared to the control and model groups, remarkable increases of fecal short-chain fatty acids were detected in both N-Asta and S-Asta pre-treatment groups, suggesting the contribution of N-Asta and S-Asta adsorption to SCFA-producing bacteria enrichment. Furthermore, the genera of Ruminococcaceae_uncultured, Ruminiclostridium_9, Ruminococcaceae_unclassified and Ruminococcus_1 showed high correlations with propionic acid, isobutyric acid, butyric acid, isovaleric acid and valeric acid increases in the S-Asta pre-treated group. Seventeen plasma biomarker metabolites in more than 10 metabolic pathways were responsible for the difference between the N-Asta and S-Asta pre-treated groups. Metabolites GSH, retinol, all-trans-Retinoic acid and taurine related to antioxidant activities were significantly accumulated in the S-Asta pre-treated group, while increasing taurocholic acid levels associated with the anti-inflammatory activity was found in the N-Asta-pre-treated group. Therefore, N-Asta and S-Asta could have potential applications in counterbalancing intestinal flora and metabolite disturbances by overdose chemical induction.


Acetaminophen/adverse effects , Gastrointestinal Microbiome/drug effects , Metabolome/drug effects , Oxidative Stress/drug effects , Animals , Antioxidants/pharmacology , Disease Models, Animal , Male , Metabolomics , Rats , Rats, Sprague-Dawley , Xanthophylls/pharmacology
19.
Biochem J ; 479(3): 425-444, 2022 02 11.
Article En | MEDLINE | ID: mdl-35048967

There has been a concern that sodium-glucose cotransporter 2 (SGLT2) inhibitors could reduce skeletal muscle mass and function. Here, we examine the effect of canagliflozin (CANA), an SGLT2 inhibitor, on slow and fast muscles from nondiabetic C57BL/6J mice. In this study, mice were fed with or without CANA under ad libitum feeding, and then evaluated for metabolic valuables as well as slow and fast muscle mass and function. We also examined the effect of CANA on gene expressions and metabolites in slow and fast muscles. During SGLT2 inhibition, fast muscle function is increased, as accompanied by increased food intake, whereas slow muscle function is unaffected, although slow and fast muscle mass is maintained. When the amount of food in CANA-treated mice is adjusted to that in vehicle-treated mice, fast muscle mass and function are reduced, but slow muscle was unaffected during SGLT2 inhibition. In metabolome analysis, glycolytic metabolites and ATP are increased in fast muscle, whereas glycolytic metabolites are reduced but ATP is maintained in slow muscle during SGLT2 inhibition. Amino acids and free fatty acids are increased in slow muscle, but unchanged in fast muscle during SGLT2 inhibition. The metabolic effects on slow and fast muscles are exaggerated when food intake is restricted. This study demonstrates the differential effects of an SGLT2 inhibitor on slow and fast muscles independent of impaired glucose metabolism, thereby providing new insights into how they should be used in patients with diabetes, who are at a high risk of sarcopenia.


Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Slow-Twitch/drug effects , Adenosine Triphosphate/metabolism , Adenylate Kinase/biosynthesis , Adenylate Kinase/genetics , Adipose Tissue, White/drug effects , Amino Acids/metabolism , Animals , Body Weight/drug effects , Canagliflozin/pharmacology , Eating/drug effects , Fatty Acids, Nonesterified/metabolism , Gene Expression Regulation/drug effects , Gene Ontology , Glycolysis , Hand Strength , Liver/drug effects , Male , Metabolome/drug effects , Mice , Mice, Inbred C57BL , Muscle Fibers, Fast-Twitch/metabolism , Muscle, Skeletal/drug effects , Organ Size/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sodium-Glucose Transporter 2/physiology , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , TOR Serine-Threonine Kinases/biosynthesis , TOR Serine-Threonine Kinases/genetics
20.
Dev Cell ; 57(3): 361-372.e5, 2022 02 07.
Article En | MEDLINE | ID: mdl-35045336

The symbiotic relationship between commensal microbes and host animals predicts unidentified beneficial impacts of individual bacterial metabolites on animal physiology. Peptidoglycan fragments (muropeptides) from the bacterial cell wall are known for their roles in pathogenicity and for inducing host immune responses. However, the potential beneficial usage of muropeptides from commensal bacteria by the host needs exploration. We identified a striking role for muropeptides in supporting mitochondrial homeostasis, development, and behaviors in Caenorhabditis elegans. We determined that the beneficial molecules are disaccharide muropeptides containing a short AA chain, and they enter intestinal-cell mitochondria to repress oxidative stress. Further analyses indicate that muropeptides execute this role by binding to and promoting the activity of ATP synthase. Therefore, given the exceptional structural conservation of ATP synthase, the role of muropeptides as a rare agonist of the ATP synthase presents a major conceptual modification regarding the impact of bacterial cell metabolites on animal physiology.


ATP Synthetase Complexes/metabolism , Caenorhabditis elegans/physiology , Homeostasis , Mitochondria/metabolism , Peptides/metabolism , Peptidoglycan/metabolism , Animals , Enzyme Inhibitors/pharmacology , Escherichia coli/metabolism , Feeding Behavior/drug effects , HEK293 Cells , Humans , Intestines/metabolism , Metabolome/drug effects , Mitochondria/drug effects , Models, Biological , Oxidative Stress/drug effects , Stress, Physiological/drug effects
...