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










Publication year range
1.
J Obstet Gynaecol Res ; 50(6): 1010-1019, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38576101

ABSTRACT

BACKGROUND: Ovarian cancer (OVCA) stands as one of the most fatal gynecological malignancies. Honokiol (HNK) has been substantiated by numerous studies for its anti-tumor activity against malignancies including OVCA. Consequently, this work was designed to elucidate the impact of HNK-mediated modulation of the YAP/TAZ pathway on the biological functions of OVCA cells. METHODS: OVCA cells were subjected to treatment with varying concentrations (0, 25, 50, 75, and 100 µM) of HNK, concomitant with the administration of YAP agonist (XMU). Assessment of cellular viability was executed employing the CCK-8 assay, while quantification of cellular proliferation transpired via colony formation assays. Apoptosis was ascertained using flow cytometry, and expression of apoptosis-related proteins (caspase-3, Bcl-2, Bax), EMT-related proteins (E-cadherin, N-cadherin), migration-associated proteins (MMP-2, MMP-9), and YAP/TAZ pathway-related proteins was evaluated by western blot. Transwell experiments were conducted to assess cellular migratory and invasive propensities. Xenograft tumor models were built to observe tumor growth (volume and weight), apoptosis was assessed by TUNEL staining, and Ki67 expression was evaluated through IHC. RESULTS: HNK exerted inhibitory effects on the viability and proliferative capacity of OVCA cells, elicited apoptotic responses, curtailed the migratory and invasive tendencies of cells, and downregulated the YAP/TAZ pathway. Stimulation with YAP agonist (XMU-MP-1) partially attenuated the impacts of HNK on OVCA cell biology. Experiments in vivo confirmed that HNK inhibited OVCA tumor growth. CONCLUSION: The outcomes of this investigation conclusively established that HNK orchestrated the modulation of the YAP/TAZ pathway, thereby exerting control over the malignant phenotypic manifestations of OVCA cells. The ascertained function of HNK in restraining cellular proliferation and tumor progression provided novel evidence of its anti-proliferative activity within OVCA cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Biphenyl Compounds , Lignans , Ovarian Neoplasms , Transcription Factors , YAP-Signaling Proteins , Female , Humans , Lignans/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Biphenyl Compounds/pharmacology , Transcription Factors/metabolism , Cell Line, Tumor , Animals , Adaptor Proteins, Signal Transducing/metabolism , Mice , Apoptosis/drug effects , Cell Proliferation/drug effects , Signal Transduction/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Cell Movement/drug effects , Xenograft Model Antitumor Assays , Mice, Nude , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Allyl Compounds , Phenols
2.
J Obstet Gynaecol Res ; 50(5): 864-872, 2024 May.
Article in English | MEDLINE | ID: mdl-38480480

ABSTRACT

BACKGROUND: Ovarian cancer (OVCA) is prevalent in female reproductive organs. Despite recent advances, clinical outcomes remain poor, warranting fresh treatment avenues. Honokiol has an inhibitory effect on proliferation, invasion, and survival of cancer cells in vitro and in vivo. Therefore, this study intended to explore specific molecular mechanism by which honokiol affected OVCA progression. METHODS: Bioinformatics analyzed the drug honokiol that bound to OTU deubiquitinase, ubiquitin aldehyde binding 2 (OTUB2). Cellular thermal shift assay (CETSA) verified the binding relationship between honokiol and OTUB2. Cell counting kit 8 (CCK-8) tested the IC50 value and cell viability of OVCA cells after honokiol treatment. Corresponding assay kits determined malonic dialdehyde (MDA) and Fe2+ levels in OVCA cells. Flow cytometry measured reactive oxygen species levels. Western blot detected OTUB2, SLC7A11, and transcriptional co-activators Yes-associated protein (YAP) expression, and quantitative polymerase chain reaction (qPCR) detected OTUB2 expression. Immunohistochemistry (IHC) detected the expression level of Ki67 protein in tumor tissues. RESULTS: Honokiol was capable of inducing ferroptosis in OVCA cells. CETSA confirmed that honokiol could bind to OTUB2. Further cell functional and molecular experiments revealed that honokiol induced ferroptosis in OVCA cells via repression of YAP signaling pathway through binding to OTUB2. In addition, in vivo experiments have confirmed that honokiol could inhibit the growth of OVCA. CONCLUSION: Honokiol induced ferroptosis in OVCA cells via repression of YAP signaling pathway through binding to OTUB2, implicating that OTUB2 may be an effective target for OVCA treatment, and our study results may provide new directions for development of more effective OVCA treatment strategies.


Subject(s)
Allyl Compounds , Biphenyl Compounds , Ferroptosis , Lignans , Ovarian Neoplasms , Phenols , Humans , Female , Lignans/pharmacology , Ferroptosis/drug effects , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Transcription Factors/metabolism , YAP-Signaling Proteins/metabolism , Deubiquitinating Enzymes/metabolism , Adaptor Proteins, Signal Transducing/metabolism
3.
Appl Environ Microbiol ; 90(4): e0000724, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38501861

ABSTRACT

With its estrogenic activity, (S)-equol plays an important role in maintaining host health and preventing estrogen-related diseases. Exclusive production occurs through the transformation of soy isoflavones by intestinal bacteria, but the reasons for variations in (S)-equol production among different individuals and species remain unclear. Here, fecal samples from humans, pigs, chickens, mice, and rats were used as research objects. The concentrations of (S)-equol, along with the genetic homology and evolutionary relationships of (S)-equol production-related genes [daidzein reductase (DZNR), daidzein racemase (DDRC), dihydrodaidzein reductase (DHDR), tetrahydrodaidzein reductase (THDR)], were analyzed. Additionally, in vitro functional verification of the newly identified DDRC gene was conducted. It was found that approximately 40% of human samples contained (S)-equol, whereas 100% of samples from other species contained (S)-equol. However, there were significant variations in (S)-equol content among the different species: rats > pigs > chickens > mice > humans. The distributions of the four genes displayed species-specific patterns. High detection rates across various species were exhibited by DHDR, THDR, and DDRC. In contrast, substantial variations in detection rates among different species and individuals were observed with respect to DZNR. It appears that various types of DZNR may be associated with different concentrations of (S)-equol, which potentially correspond to the regulatory role during (S)-equol synthesis. This enhances our understanding of individual variations in (S)-equol production and their connection with functional genes in vitro. Moreover, the newly identified DDRC exhibits higher potential for (S)-equol synthesis compared to the known DDRC, providing valuable resources for advancing in vitro (S)-equol production. IMPORTANCE: (S)-equol ((S)-EQ) plays a crucial role in maintaining human health, along with its known capacity to prevent and treat various diseases, including cardiovascular diseases, metabolic syndromes, osteoporosis, diabetes, brain-related diseases, high blood pressure, hyperlipidemia, obesity, and inflammation. However, factors affecting individual variations in (S)-EQ production and the underlying regulatory mechanisms remain elusive. This study examines the association between functional genes and (S)-EQ production, highlighting a potential correlation between the DZNR gene and (S)-EQ content. Various types of DZNR may be linked to the regulation of (S)-EQ synthesis. Furthermore, the identification of a new DDRC gene offers promising prospects for enhancing in vitro (S)-EQ production.


Subject(s)
Equol , Isoflavones , Animals , Humans , Mice , Rats , Swine , Equol/genetics , Equol/metabolism , Racemases and Epimerases , Chickens/metabolism , Isoflavones/metabolism , Oxidoreductases/metabolism
4.
Soft Matter ; 20(1): 192-200, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38073481

ABSTRACT

Biofilms are initially formed by substances such as proteins secreted by bacteria adhering to a surface. To achieve a durable antibacterial material, biodegradable dihydroxyl-terminated poly[(ethylene oxide)-co-(ethylene carbonate)] (PEOC(OH)2) with anti-protein adsorption properties was synthesized in this study. Further polycondensation of PEOC(OH)2 and isophorone diisocyanate (IPDI) led to biodegradable polyurethane (PEOC-PU) with PEOC as the soft segment. For comparison, polyurethanes with polyethylene glycol (PEG-PU) and polypropylene glycol (PPG-PU) as soft segments were also synthesized. The chemical structures of the polyurethanes were characterized by 1H NMR and FTIR. The biodegradation behavior of PEOC-PU promoted by lipase due to the presence of ethylene carbonate units was also studied. Their resistance to proteins was studied using quartz crystal microbalance with dissipation (QCM-D) and the results revealed that PEOC-PU exhibited excellent nonspecific resistance to proteins. The colonization of microorganisms on PU in the liquid culture medium was further examined and the results showed that PEOC-PU exhibited excellent antibacterial adhesion performance due to its protein resistance and biodegradation.


Subject(s)
Polyurethanes , Proteins , Polyurethanes/chemistry , Adsorption , Polyethylene Glycols/chemistry , Anti-Bacterial Agents , Biocompatible Materials/chemistry
5.
Lett Appl Microbiol ; 76(12)2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38031336

ABSTRACT

The gut microbiota consists of a vast and diverse assemblage of microorganisms that play a pivotal role in maintaining host health. Nevertheless, a significant portion of the human gut microbiota remains uncultivated. Plasmids, a type of MGE, assume a critical function in the biological evolution and adaptation of bacteria to varying environments. To investigate the plasmids present within the gut microbiota community, we used the transposon-aided capture method (TRACA) to explore plasmids derived from the gut microbiota. In this study, fecal samples were collected from two healthy human volunteers and subsequently subjected to the TRACA method for plasmid isolation. Then, the complete sequence of the plasmids was obtained using the genome walking method, and sequence identity was also analyzed. A total of 15 plasmids were isolated. At last, 13 plasmids were successfully sequenced, of which 12 plasmids were highly identical to the plasmids in the National Center for Biotechnology Information (NCBI) database and were all small plasmids. Furthermore, a putative novel plasmid, named pMRPHD, was isolated, which had mobilized elements (oriT and oriV) and a potential type II restriction-modification (R-M) system encoded by DNA cytosine methyltransferase and type II restriction enzyme (Ban I), whose specific functions and applications warrant further exploration.


Subject(s)
Bacteria , Humans , Plasmids/genetics , Bacteria/genetics
6.
Acta Biochim Biophys Sin (Shanghai) ; 55(5): 783-794, 2023 May 25.
Article in English | MEDLINE | ID: mdl-37232573

ABSTRACT

Bone marrow mesenchymal stem cell (BMSC)-derived exosomes are a promising therapeutic agent for human disease, but their effects on neural stem cells (NSCs) subject to spinal cord ischaemia-reperfusion injury (SCIRI) remain unknown. Here, we examine the impact of miR-199a-5p-enriched exosomes derived from BMSCs on NSC proliferation. We establish a rat model of aortic cross-clamping to induce SCIRI in vivo and a primary NSC model of oxygen-glucose deprivation/reoxygenation (OGD/R) to simulate SCIRI in vitro. CCK8, EdU, and BrdU assays are performed to evaluate the proliferation of NSCs. Hematoxylin and eosin (H&E) staining is used to determine the number of surviving neurons. The Basso, Beattie, and Bresnahan (BBB) scale and inclined plane test (IPT) are used to evaluate hind limb motor function. DiO-labelled exosomes are efficiently internalized by NSCs and increase ectopic amounts of miR-199a-5p, which promotes the proliferation of NSCs. In contrast, exosomes derived from miR-199a-5p-depleted BMSCs exert fewer beneficial effects. MiR-199a-5p targets and negatively regulates glycogen synthase kinase 3ß (GSK-3ß) and increases nuclear ß-catenin and cyclin D1 levels. miR-199a-5p inhibition reduces the total number of EdU-positive NSCs after OGD/R, but the GSK-3ß inhibitor CHIR-99021 reverses this effect. In vivo, intrathecal injection of BMSC-derived exosomes increases the proliferation of endogenous spinal cord NSCs after SCIRI. In addition, more proliferating NSCs are found in rats intrathecally injected with exosomes overexpressing miR-199a-5p. In summary, miR-199a-5p in BMSC-derived exosomes promotes NSC proliferation via GSK-3ß/ß-catenin signaling.


Subject(s)
Exosomes , Mesenchymal Stem Cells , MicroRNAs , Neural Stem Cells , Reperfusion Injury , Rats , Humans , Animals , MicroRNAs/genetics , beta Catenin/genetics , Glycogen Synthase Kinase 3 beta/genetics , Exosomes/genetics , Cell Proliferation
7.
FASEB J ; 37(2): e22760, 2023 02.
Article in English | MEDLINE | ID: mdl-36607643

ABSTRACT

The activation of endogenous neural stem cells (NSCs) is considered an important mechanism of neural repair after mechanical spinal cord injury; however, whether endogenous NSC proliferation can also occur after spinal cord ischemia-reperfusion injury (SCIRI) remains unclear. In this study, we aimed to verify the existence of endogenous NSC proliferation after SCIRI and explore the underlying molecular mechanism. NSC proliferation was observed after SCIRI in vivo and oxygen-glucose deprivation and reperfusion (OGD/R) in vitro, accompanied by a decrease in forkhead box protein O 3a (FOXO3a) expression. This downward trend was regulated by the increased expression of microRNA-872-5p (miR-872-5p). miR-872-5p affected NSC proliferation by targeting FOXO3a to increase the expression of ß-catenin and T-cell factor 4 (TCF4). In addition, TCF4 in turn acted as a transcription factor to increase the expression level of miR-872-5p, and knockdown of FOXO3a enhanced the binding of TCF4 to the miR-872-5p promoter. In conclusion, SCIRI in vivo and OGD/R in vitro stimulated the miR-872-5p/FOXO3a/ß-catenin-TCF4 pathway, thereby promoting NSC proliferation. At the same time, FOXO3a affected TCF4 transcription factor activity and miR-872-5p expression, forming a positive feedback loop that promotes NSC proliferation.


Subject(s)
MicroRNAs , Neural Stem Cells , Reperfusion Injury , Rats , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism , Neural Stem Cells/metabolism , Spinal Cord/metabolism , Oxygen/metabolism , Cell Proliferation , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Apoptosis
8.
Sheng Wu Gong Cheng Xue Bao ; 38(10): 3682-3694, 2022 Oct 25.
Article in Chinese | MEDLINE | ID: mdl-36305403

ABSTRACT

Genome sequences of 4 644 representative strains from human gut microbiota were analyzed to mine gene clusters for biosynthesis of novel secondary metabolites, as well as genes encoding antibiotic resistance and virulence factors. AntiSMASH analysis showed that more than 60% of the representative strains encoded at least one secondary metabolite gene cluster, and 8 potential novel secondary metabolite gene clusters were identified from 8 unculturable bacteria. The secondary metabolite gene clusters in human intestine are mainly composed of nonribosomal peptide synthetase (NRPS), bacteriocin, arylpolyene, terpene, betalactone and NRPS like gene clusters distributed in Clostridia, Bacilli, Gammaproteobacteria, Bacteroidia, Actinobacteria and Negativicutes. PathoFact analysis showed that genes encoding antibiotic resistance and virulence factors are widely distributed in representative strains, but the frequency encoded by potential pathogens is significantly higher than that of non-potential pathogens. The frequency of genes encoding secretory toxins such as outer membrane protein, PapC N-terminal domain, PapC C-terminal domain, peptidase M16 inactive domain, and non-secretory toxins such as nitroreductase family, AcrB/AcrD/AcrF family, PLD-like domain, Cupin domain, putative hemolysin, S24-like peptidase, phosphotransferase enzyme family, endonuclease/ exonuclease/ phosphatase family, glyoxalase/ bleomycin resistance was high in potential pathogens. This study may facilitate mining new microbial natural products from the intestinal microbiome, understanding the colonization and infection mechanism of intestinal microorganisms, and providing targeted prevention and treatment of intestinal microbial related diseases.


Subject(s)
Bacteria , Multigene Family , Humans , Virulence , Drug Resistance, Microbial , Virulence Factors , Peptide Hydrolases
9.
J Clin Lab Anal ; 36(8): e25470, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35778954

ABSTRACT

BACKGROUND: Serous ovarian carcinoma (SOC) is a common malignant tumor in female reproductive system. Long noncoding RNA (lncRNA) LIFR-AS1 is a tumor suppressor gene in colorectal cancer, but its effect and underlying mechanism in SOC are still unclear. Therefore, this study focuses on unveiling the regulatory mechanism of LIFR-AS1 in SOC. METHODS: The relationship between LIFR-AS1 expression and prognosis of SOC patients was analyzed by TCGA database and Starbase, and then, the LIFR-AS1 expression in SOC tissues and cells was detected by quantitative real-time PCR (qRT-PCR) and in situ hybridization (ISH). Besides, the relationship between LIFR-AS1 and clinical characteristics was analyzed. Also, the effects of LIFR-AS1 on the biological behaviors of SOC cells were measured by Cell Counting Kit-8, colony formation, and wound-healing and Transwell assays, respectively. Western blot and qRT-PCR were employed to determine the protein expressions of genes related to proliferation (PCNA), apoptosis (cleaved caspase-3), epithelial-mesenchymal transition (E-cadherin, N-cadherin, and Snail). RESULTS: LIFR-AS1 was lowly expressed in SOC, which was correlated with the poor prognosis of SOC patients. Low expression of LIFR-AS1 in SOC was associated with the tumor size, clinical stage, lymph node metastasis, and distant metastasis. LIFR-AS1 overexpression promoted the expressions of cleaved caspase-3 and E-cadherin while suppressing the malignant behaviors (proliferation, migration, and invasion) of SOC cells, the expressions of PCNA, N-cadherin, and Snail. Besides, silencing LIFR-AS1 exerted the effects opposite to overexpressed LIFR-AS1. CONCLUSION: LIFR-AS1 overexpression inhibits biological behaviors of SOC cells, which may be a new therapeutic method.


Subject(s)
Ovarian Neoplasms , RNA, Long Noncoding , Cadherins , Carcinoma, Ovarian Epithelial/genetics , Caspase 3/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proliferating Cell Nuclear Antigen/genetics , RNA, Long Noncoding/genetics
10.
Front Microbiol ; 13: 901745, 2022.
Article in English | MEDLINE | ID: mdl-35668767

ABSTRACT

(S)-equol (EQ) is an isoflavone with high estrogen-like activity in the human body, and is only produced by some gut bacteria in vivo. It plays an important role in maintaining individual health, however, the dearth of resources associated with (S)-EQ-producing bacteria has seriously restricted the production and application of (S)-EQ. We report here a new functional gene KEC48-07020 (K-07020) that was identified from a chick (S)-EQ-producing bacterium (Clostridium sp. ZJ6, ZJ6). We found that recombinant protein of K-07020 possessed similar function to daidzein reductase (DZNR), which can convert daidzein (DZN) into R/S-dihydrodaidzein (R/S-DHD). Interestingly, K-07020 can reversely convert (R/S)-DHD (DHD oxidase) into DZN even without cofactors under aerobic conditions. Additionally, high concentrations of (S)-EQ can directly promote DHD oxidase but inhibit DZNR activity. Molecular docking and site-directed mutagenesis revealed that the amino acid > Arg75 was the active site of DHD oxidase. Subsequently, an engineered E. coli strain based on K-07020 was constructed and showed higher yield of (S)-EQ than the engineered bacteria from our previous work. Metagenomics analysis and PCR detection surprisingly revealed that K-07020 and related bacteria may be prevalent in the gut of humans and animals. Overall, a new DZNR from ZJ6 was found and identified in this study, and its bidirectional enzyme activities and wide distribution in the gut of humans and animals provide alternative strategies for revealing the individual regulatory mechanisms of (S)-EQ-producing bacteria.

11.
Appl Environ Microbiol ; 88(10): e0041022, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35477272

ABSTRACT

Antibiotic resistance is a serious medical issue driven by antibiotic misuse. Bifidobacteria may serve as a reservoir for antibiotic resistance genes (ARGs) that have the potential risk of transfer to pathogens. The erythromycin resistance gene erm(X) is an ARG with high abundance in bifidobacteria, especially in Bifidobacterium longum species. However, the characteristics of the spread and integration of the gene erm(X) into the bifidobacteria genome are poorly understood. In this study, 10 tetW-positive bifidobacterial strains and 1 erm(X)-positive bifidobacterial strain were used to investigate the transfer of ARGs. Conjugation assays found that the erm(X) gene could transfer to five other bifidobacterial strains. Dimethyl sulfoxide (DMSO) and vorinostat significantly promoted the transfer of the erm(X) from strain Bifidobacterium catenulatum subsp. kashiwanohense DSM 21854 to Bifidobacterium longum subsp. suis DSM 20211. Whole-genome sequencing and comparative genomic analysis revealed that the erm(X) gene was located on the genomic island BKGI1 and that BKGI1 was conjugally mobile and transferable. To our knowledge, this is the first report that a genomic island-mediated gene erm(X) transfer in bifidobacteria. Additionally, BKGI1 is very unstable in B. catenulatum subsp. kashiwanohense DSM 21854 and transconjugant D2TC and is highly excisable and has an intermediate circular formation. In silico analysis showed that the BKGI1 homologs were also present in other bifidobacterial strains and were especially abundant in B. longum strains. Thus, our results confirmed that genomic island BKGI1 was one of the vehicles for erm(X) spread. These findings suggest that genomic islands play an important role in the dissemination of the gene erm(X) among Bifidobacterium species. IMPORTANCE Bifidobacteria are a very important group of gut microbiota, and the presence of these bacteria has many beneficial effects for the host. Thus, bifidobacteria have attracted growing interest owing to their potential probiotic properties. Bifidobacteria have been widely exploited by the food industry as probiotic microorganisms, and some species have a long history of safe use in food and feed production. However, the presence of antibiotic resistance raises the risk of its application. In this study, we analyzed the transfer of the erythromycin resistance gene erm(X) and revealed that the molecular mechanism behind the spread of the gene erm(X) was mediated by genomic island BKGI1. To the best of our knowledge this is the first report to describe the transfer of the gene erm(X) via genomic islands among bifidobacteria. This may be an important way to disseminate the gene erm(X) among bifidobacteria.


Subject(s)
Erythromycin , Genomic Islands , Anti-Bacterial Agents/pharmacology , Bifidobacterium/genetics , Erythromycin/pharmacology
12.
Front Microbiol ; 12: 634204, 2021.
Article in English | MEDLINE | ID: mdl-33679666

ABSTRACT

A stable intestinal microflora is an essential prerequisite for human health. This study investigated the interaction between Escherichia coli exopolysaccharides (named EPS-m2) and the human gut microbiota (HGM) in vitro. The EPS-m2 was produced by E. coli WM3064 when treated with ceftriaxone. The monosaccharide composition analysis revealed that EPS-m2 is composed of glucuronic acid, glucose, fucose, galactose/N-acetyl glucosamine, arabinose, xylose, and ribose with a molar ratio of approximately 77:44:29:28:2:1:1. The carbohydrates, protein, and uronic acids contents in EPS-m2 was 78.6 ± 0.1%, 4.38 ± 0.11%, and 3.86 ± 0.09%, respectively. In vitro batch fermentation experiments showed that 77% of EPS-m2 could be degraded by human fecal microbiota after 72 h of fermentation. In reverse, 16S rRNA gene sequencing analysis showed that EPS-m2 increased the abundance of Alistipes, Acinetobacter, Alloprevotella, Howardella, and Oxalobacter; GC detection illustrated that EPS-m2 enhanced the production of SCFAs. These findings indicated that EPS-m2 supplementation could regulate the HGM and might facilitate modulation of human health.

13.
Curr Microbiol ; 77(12): 4104-4113, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33057753

ABSTRACT

Bifidobacteria are typical commensals inhabiting the human intestine and are beneficial to the host because of their probiotic properties. One of the risks concerning probiotics is the potential of introducing antibiotic resistance genes (ARGs) to the host gut pathogens. This study was aimed to depict the general antibiotic resistance characteristics of the genus Bifidobacterium by combining the reported phenotype dataset and in silico genotype prediction. Bifidobacteria were mostly reported to be sensitive to beta-lactams, glycopeptides, chloramphenicol, and rifampicin, but resistant to aminoglycosides, polypeptides, quinolones, and mupirocin. Generally, the resistance phenotypes to erythromycin, tetracycline, fusidic acid, metronidazole, clindamycin, and trimethoprim were variable. Besides cmX and tetQ, characterized in bifidobacterial resident plasmids, 3520 putative ARGs were identified from 831 bifidobacterial genomes through BLASTP search. The identified ARGs matched thirty-eight reference ARGs, four of which seemed to be mutant housekeeping genes. The two high-abundant ARGs, tetW and ermX, were found to have different distribution traits. The predicted ARGs reasonably explained most of the corresponding resistant phenotypes in the published literature.


Subject(s)
Anti-Bacterial Agents , Bifidobacterium , Anti-Bacterial Agents/pharmacology , Bifidobacterium/genetics , Computer Simulation , Drug Resistance, Microbial , Genes, Bacterial , Genotype , Humans , Phenotype
14.
PLoS One ; 15(3): e0230200, 2020.
Article in English | MEDLINE | ID: mdl-32214324

ABSTRACT

Statins, a class of drugs that can effectively remove cholesterol from serum, are used to regulate plasma total cholesterol and reduce the risk of cardiovascular diseases, but it is still unclear whether the drug are modulated by gut microbiota or the structures of gut microbiota are shaped by statins. We investigated the interactions between statins and the human gut microbiota during the in vitro fermentation process by 16S rRNA gene sequencing, gas chromatography (GC), and high-performance liquid chromatography (HPLC) analyses. The presence of fluvastatin (FLU2) specifically promoted the growth of Escherichia/Shigella, Ruminococcaceae UCG 014, and Sutterella. However, the composition of the gut bacterial microbiota remained relatively static in samples treated with rosuvastatin (ROS), simvastatin (SIM), and atorvastatin (ATO). The PICRUSt program predicted moderate differences in the functional categories related to the biosynthesis of other secondary metabolites, cellular processes and signaling, and signal transduction in the FLU2 fermentation samples. Our study revealed substantial variation in the structure and function of microbiomes from the FLU2-treated samples. In addition, short-chain fatty acids (SCFAs) were also significantly decreased in FLU2-treated samples compared with the samples treated with other stains. Statins can be degraded by the human gut microbiota in vitro, and the degradation rate was approximately 7%-30% and 19%-48% after fermentation was allowed to proceed for 24 h and 48 h, respectively. Generally, FLU2 could largely shape the composition and function of human gut microbiota, which resulted in changes in the production of SCFAs. In turn, all statins could be degraded or modified by the gut microbiota. Our study paves the way for elucidating statin-gut microbiota interactions in vitro towards the improvement of the host health and personalized medicine.


Subject(s)
Bacteria/drug effects , Gastrointestinal Microbiome/drug effects , Microbiota/drug effects , Adolescent , Adult , Bacteria/genetics , Cardiovascular Diseases/drug therapy , Fatty Acids, Volatile/genetics , Feces/microbiology , Female , Fermentation/drug effects , Fermentation/genetics , Gastrointestinal Microbiome/genetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Male , Microbiota/genetics , RNA, Ribosomal, 16S/genetics , Young Adult
15.
Int J Biol Macromol ; 150: 991-999, 2020 May 01.
Article in English | MEDLINE | ID: mdl-31751742

ABSTRACT

In this study, an exopolysaccharide (EPS) named EPS-RB was produced when the gene cluster ycjD-fabI-yciW-rnb were overexpressed in E. coli. Monosaccharide composition analysis revealed that EPS-RB is a novel EPS that consisted of L-fucose, L-arabinose, D-galactose/N-acetyl glucosamine, D-glucose, D-xylose, D-ribose, and D-glucuronic acid, and their molecular ratio was approximately 80:3:53:69:1:2:64. The content of carbohydrates, protein, and uronic acids in EPS-RB was 90.35 ± 1.35%, 2.62 ± 0.05% and 8.16 ± 1.00%, respectively. The interaction between EPS-RB and gut microbiota was investigated using an in vitro batch fermentation system. The results showed that ~96% of EPS-RB can be degraded by human fecal microbiota after 72 h fermentation, but few can be degraded by mouse cecal microbiota. Furthermore, high-throughput sequencing showed that EPS-RB regulates the human gut microbiota. The genera Collinsella, Butyricimonas, and Hafnia were enriched in group VIR (EPS-RB as a carbon source) when compared with group VI (no carbon source) and VIS (starch as a carbon source). Short-chain fatty acids (SCFAs) production analysis showed that their concentration was significantly higher in group VIR than groups VI and VIS after 72 h fermentation. In summary, an EPS-RB in E. coli was isolated and its regulatory function on gut microbiota was analyzed.


Subject(s)
Escherichia coli/metabolism , Gastrointestinal Microbiome/drug effects , Polysaccharides, Bacterial/pharmacology , Animals , Bacteria/classification , Bacteria/drug effects , Batch Cell Culture Techniques , Fatty Acids, Volatile , Feces/microbiology , Fermentation , High-Throughput Screening Assays , Humans , Mice , Monosaccharides/analysis , Starch
16.
J Vis Exp ; (150)2019 08 23.
Article in English | MEDLINE | ID: mdl-31498314

ABSTRACT

Human intestinal microorganisms have recently become an important target of research in promoting human health and preventing diseases. Consequently, investigations of interactions between endobiotics (e.g., drugs and prebiotics) and gut microbiota have become an important research topic. However, in vivo experiments with human volunteers are not ideal for such studies due to bioethics and economic constraints. As a result, animal models have been used to evaluate these interactions in vivo. Nevertheless, animal model studies are still limited by bioethics considerations, in addition to differing compositions and diversities of microbiota in animals vs. humans. An alternative research strategy is the use of batch fermentation experiments that allow evaluation of the interactions between endobiotics and gut microbiota in vitro. To evaluate this strategy, bifidobacterial (Bif) exopolysaccharides (EPS) were used as a representative xenobiotic. Then, the interactions between Bif EPS and human gut microbiota were investigated using several methods such as thin-layer chromatography (TLC), bacterial community compositional analysis with 16S rRNA gene high-throughput sequencing, and gas chromatography of short-chain fatty acids (SCFAs). Presented here is a protocol to investigate the interactions between endobiotics and human gut microbiota using in vitro batch fermentation systems. Importantly, this protocol can also be modified to investigate general interactions between other endobiotics and gut microbiota.


Subject(s)
Gastrointestinal Microbiome/drug effects , Prebiotics , Xenobiotics/pharmacology , Animals , Bifidobacterium/genetics , Fatty Acids, Volatile/analysis , Fermentation , Humans , Intestines/microbiology , RNA, Ribosomal, 16S
17.
Article in English | MEDLINE | ID: mdl-30533788

ABSTRACT

The surface exopolysaccharide of Bifidobacterium spp. was found to be involved in many processes, including bacterial colonization and host immune modulation. Here, we report the draft genome sequence of an exopolysaccharide-producing strain of Bifidobacterium longum, BB-79.

18.
Sheng Wu Gong Cheng Xue Bao ; 34(8): 1346-1360, 2018 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-30152220

ABSTRACT

Antimicrobial resistance is on the rise while the number of antibiotics being brought to market continues to drop. Drug-resistant genes and drug-resistant bacteria infection have seriously threatened human health. Therefore, antimicrobial resistance presents an ongoing challenge that requires multifaceted approaches including: biomedical innovation; improved surveillance of antibiotic consumption and antimicrobial resistance generated rates; prevention of health-care-associated infections and transmission of multidrug-resistant bacteria and environmental dissemination; rapid microbiological diagnosis; and curtailed clinical and veterinary misuse. Fortunately, combating antimicrobial resistance has been highly valued and supported by the government, scientists and entrepreneurs of various countries. With the continuous introduction of new technologies, new products, and new management measures, the problem of antimicrobial resistance must be controlled and alleviated.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria , Drug Resistance, Bacterial , Humans
19.
J Biomed Nanotechnol ; 14(3): 601-608, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29663932

ABSTRACT

Antibiotic resistance is a growing threat to human health. More than anything else, multidrug-resistant gram-negative bacteria pose the biggest challenge to healthcare, which is predominantly due to the lack of effective therapeutic options. In the present study, we investigated the activities of peptides HX-12A, HX-12B and HX-12C against MDRAB and MDRKP isolates in vitro. Those three peptides displayed high antibacterial activities and rapid bactericidal effects against MDRAB and MDRKP isolates. Additionally, the peptides retained their activity even being present in pH 6.8 and 8.0, salt (NaCl 100 mM, CaCl2 1 mM, MgCl2 1 mM) or human serum (5%), especially peptide HX-12C. Moreover, the killing kinetics and transmission electron microscopy results suggested that the possible sterilization mechanism of peptides is to destroy the cell membrane, which makes it more difficult for bacteria to develop resistance. Thus, these three peptides may be developed into new antimicrobial agents.


Subject(s)
Drug Resistance, Bacterial , Anti-Bacterial Agents , Microbial Sensitivity Tests
20.
Int J Med Microbiol ; 306(8): 686-696, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27600408

ABSTRACT

The CRISPR-Cas (clustered regularly interspaced short palindromic repeats [CRISPR]-CRISPR associated proteins [Cas]) system can provide prokaryote with immunity against invading mobile genetic elements (MGEs) such as phages and plasmids, which are the main sources of staphylococcal accessory genes. To date, only a few Staphylococcus aureus strains containing CRISPR-Cas systems have been identified, but no functional study in these strains has been reported. In this study, 6 clinical isolates of S. aureus with type III-A CRISPR-Cas systems were identified, and whole-genome sequencing and functional study were conducted subsequently. Genome sequence analysis revealed a close linkage between the CRISPR-Cas system and the staphylococcal cassette chromosome mec (SCCmec) element in five strains. Comparative sequence analysis showed that the type III-A repeats are conserved within staphylococci, despite of the decreased conservation in trailer-end repeats. Highly homologous sequences of some spacers were identified in staphylococcal MGEs, and partially complementary sequences of spacers were mostly found in the coding strand of lytic regions in staphylococcal phages. Transformation experiments showed that S. aureus type III-A CRISPR-Cas system can specifically prevent plasmid transfer in a transcription-dependent manner. Base paring between crRNA and target sequence, the endoribonuclease, and the Csm complex were proved to be necessary for type III-A CRISPR-Cas immunity.


Subject(s)
CRISPR-Cas Systems , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Gene Order , Gene Transfer, Horizontal , Genome, Bacterial , Humans , Sequence Analysis, DNA , Staphylococcal Infections/microbiology , Staphylococcus aureus/isolation & purification , Transcription, Genetic , Transformation, Bacterial
SELECTION OF CITATIONS
SEARCH DETAIL
...