1.Mechanism analysis of platelet activation induced by V. vulnificus hemolysin.
Yan WANG ; Zihan FENG ; Yaru WANG ; Shiqing LI ; Xin CHEN ; Jinglin WANG ; Yuan YUAN
Chinese Journal of Cellular and Molecular Immunology 2025;41(2):134-142
Objective To evaluate whether Vibrio vulnificus secreted exotoxin-hemolysin (VVH) can activate platelet, an important blood immune cell, and to explore the possible molecular mechanism of platelet activation by VVH. Methods Transcriptomics and immunohistochemistry were used to analyze whether Vibrio vulnificus infection caused platelet activation in mice. Then, flow cytometry was used to identify whether VVH was the main stimulator of platelet activation. Naturally expressed VVH toxin was purified and prepared. The effects of extracellular and intracellular Ca2+ signal inhibitors on VVH activated platelets were evaluated by flow cytometry and Western blotting. The immune activation effect of VVH in the early stage of Vibrio vulnificus infection was analyzed in vivo. Results VVH was the main stimulator of platelet activation in Vibrio vulnificus culture supernatant. Natural VVH can induce the increase of P-selectin (CD62P) on platelet surface, the formation of platelet-neutrophil complex (PNC), and the release of platelet microvesicles. The activation mechanism may be related to the VVH pore-dependent Ca2+-calmodulin (CaM) -myosin light chain kinase (MLCK) signaling pathway, which led to the release of platelet alpha particles and cascade activation of platelets. In a mouse model of ALD infected by Vibrio vulnificus gavage, VVH was strongly associated with platelet activation. Conclusion This study shows that VVH is an important platelet activating molecule in the early stage of Vibrio vulnificus infection, and its induction of platelet activation may be related to the pathogenic process.
Animals
;
Platelet Activation/drug effects*
;
Hemolysin Proteins/pharmacology*
;
Vibrio vulnificus/metabolism*
;
Mice
;
Blood Platelets/drug effects*
;
Vibrio Infections/immunology*
;
P-Selectin/metabolism*
;
Bacterial Proteins
;
Female
2.Mycobacterium tuberculosis PPE59 promotes its survival in host cells by regulating cytokine secretion of Mycobacterium smegmatis infected macrophages.
Chutong WANG ; Fangzheng GUO ; Yamin SONG ; Jing WEI ; Minying LI ; Hongtao WANG ; Tao XU
Chinese Journal of Cellular and Molecular Immunology 2025;41(10):875-881
Objective To study the effect of Mycobacterium tuberculosis (Mtb) Pro-Pro-Glu-59 (PPE59) protein on the biological function of Mycobacterium smegmatis (Ms) and the regulation of host cell immune response. Methods PPE59 gene fragment was obtained by PCR amplification, cloned into pALACE, constructed into recombinant pALACE-PPE59 vector, and electro-transformed into Ms. Western blot was applied to analyse PPE59 expression and subcellular localization. The survival of Ms_Vec and Ms_PPE59 under low acid (pH=3 and pH=5) conditions and active surface pressure sodium dodecyl sulfate (SDS) conditions and their intracellular survival in macrophages were analyzed. ELISA was used to detect the cytokine (IL-1β, IL-6, IL-12, TNF-α and IL-10) expression levels of Ms_Vec and Ms_PPE59 infected macrophages. Results PPE59 protein localized to the cell wall of Ms can enhance the acid-resistance and anti-SDS effect of Ms, which is conducive to the survival of Ms in macrophages. PPE59 significantly decreased the secretion levels of pro-inflammatory cytokines (IL-1β, IL-6, IL-12 and TNF-α), and promoted the secretion levels of anti-inflammatory cytokine (IL-10). Conclusion PPE59 enhances the survival ability of Ms under low acid and SDS pressure and promotes its intracellular survival by regulating the cytokine secretion levels.
Mycobacterium smegmatis/metabolism*
;
Macrophages/metabolism*
;
Cytokines/metabolism*
;
Mycobacterium tuberculosis/metabolism*
;
Bacterial Proteins/metabolism*
;
Animals
;
Mice
;
Antigens, Bacterial/metabolism*
3.Thesium chinense Turcz. alleviates antibiotic-associated diarrhea in mice by modulating gut microbiota structure and regulating the EGFR/PI3K/Akt signaling pathway.
Haonan XU ; Fang ZHANG ; Yuying HUANG ; Qisheng YAO ; Yueqin GUAN ; Hao CHEN
Journal of Southern Medical University 2025;45(2):285-295
OBJECTIVES:
To investigate the therapeutic mechanism of Thesium chinense Turcz. (TCT) for antibiotic-associated diarrhea (AAD).
METHODS:
Network pharmacology, KEGG pathway enrichment analysis and molecular docking were used to identify the shared targets and genes of TCT and AAD, the key signaling pathways and the binding between the active components in TCT and the core protein targets. In a Kunming mouse model of AAD established by intragastric administration of lincomycin hydrochloride, the effects of daily gavage of 1% carboxymethyl cellulose sodium or TCT gel solutions at 1.5 g/kg and 3 g/kg (n=10) on body weight and diarrhea were observed. HE staining, ELISA, 16S rRNA sequencing, and Western blotting were used to examine pathologies, expression levels of IL-6 and TNF-α, changes in gut microbiota, and protein expressions of EGFR, p-EGFR, PI3K, p-PI3K, Akt, and p-Akt in the colon tissues of the mice.
RESULTS:
We identified a total of 66 active components of TCT and 68 core targets including EGFR, STAT3 and PIK3CA. KEGG pathway enrichment analysis suggested that the therapeutic effects of TCT was mediated primarily through the PI3K/Akt signaling pathway. Molecular docking showed that EGFR had the highest binding affinity with coniferin, and the EGFR-coniferin complex maintained a stable conformation at 10 ns, whose stability was also confirmed by Gibbs free energy analysis. In the mouse models of AAD, treatment with TCT significantly improved colonic tissue morphology, decreased colonic levels of TNF-α and IL-6, increased gut microbiota diversity, and modulated the relative abundances of the key genera including Lactobacillus and Bacteroides. TCT treatment also markedly reduced protein expressions of p-EGFR, p-PI3K and p-Akt in the colon tissues of the mice.
CONCLUSIONS
TCT can alleviate AAD in mice by modulating gut microbiota composition, regulating the EGFR/PI3K/Akt signaling pathway, and reducing TNF‑α and IL-6 expressions.
Animals
;
Gastrointestinal Microbiome/drug effects*
;
Signal Transduction/drug effects*
;
Mice
;
ErbB Receptors/metabolism*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Diarrhea/drug therapy*
;
Phosphatidylinositol 3-Kinases/metabolism*
;
Anti-Bacterial Agents/adverse effects*
;
Drugs, Chinese Herbal/therapeutic use*
;
Molecular Docking Simulation
4.Identification of a Fusobacterial RNA-binding protein involved in host small RNA-mediated growth inhibition.
Pu-Ting DONG ; Mengdi YANG ; Jie HU ; Lujia CEN ; Peng ZHOU ; Difei XU ; Peng XIONG ; Jiahe LI ; Xuesong HE
International Journal of Oral Science 2025;17(1):48-48
Host-derived small RNAs are emerging as critical regulators in the dynamic interactions between host tissues and the microbiome, with implications for microbial pathogenesis and host defense. Among these, transfer RNA-derived small RNAs (tsRNAs) have garnered attention for their roles in modulating microbial behavior. However, the bacterial factors mediating tsRNA interaction and functionality remain poorly understood. In this study, using RNA affinity pull-down assay in combination with mass spectrometry, we identified a putative membrane-bound protein, annotated as P-type ATPase transporter (PtaT) in Fusobacterium nucleatum (Fn), which binds Fn-targeting tsRNAs in a sequence-specific manner. Through targeted mutagenesis and phenotypic characterization, we showed that in both the Fn type strain and a clinical tumor isolate, deletion of ptaT led to reduced tsRNA intake and enhanced resistance to tsRNA-induced growth inhibition. Global RNA sequencing and label-free Raman spectroscopy revealed the phenotypic differences between Fn wild type and PtaT-deficient mutant, highlighting the functional significance of PtaT in purine and pyrimidine metabolism. Furthermore, AlphaFold 3 prediction provides evidence supporting the specific binding between PtaT and Fn-targeting tsRNA. By uncovering the first RNA-binding protein in Fn implicated in growth modulation through interactions with host-derived small RNAs (sRNAs), our study offers new insights into sRNA-mediated host-pathogen interplay within the context of microbiome-host interactions.
Fusobacterium nucleatum/growth & development*
;
RNA-Binding Proteins/genetics*
;
Bacterial Proteins/genetics*
;
RNA, Bacterial/metabolism*
;
Humans
;
RNA, Transfer/metabolism*
5.Antibiotic-Depleted Lung Microbiota Modulates Surfactant Proteins Expression and Reduces Experimental Silicosis.
Qiang ZHOU ; Mei Yu CHANG ; Ning LI ; Yi GUAN ; San Qiao YAO
Biomedical and Environmental Sciences 2025;38(4):469-483
OBJECTIVE:
Recent studies have overturned the traditional concept of the lung as a "sterile organ" revealing that pulmonary microbiota dysbiosis and abnormal surfactant proteins (SPs) expression are involved in the progression of silicosis. This study aimed to investigate the relationship between abnormal SPs expression and dysbiosis of lung microbiota in silica-induced lung fibrosis, providing insights into mechanisms of silicosis.
METHODS:
Lung pathology, SPs expression, and microbiota composition were evaluated in silica-exposed mice. A mouse model of antibiotic-induced microbiota depletion was established, and alveolar structure and SPs expression were assessed. The roles of the lung microbiota and SPs in silicosis progression were further evaluated in mice with antibiotic-induced microbiota depletion, both with and without silica exposure.
RESULTS:
Silica exposure induced lung inflammation and fibrosis, along with increased expression of SP-A expression. Antibiotics (Abx)-induced microbiota depletion elevated SP-A and SP-D expression. Furthermore, silica exposure altered lung microbiota composition, enriching potentially pathogenic taxa. However, antibiotic-induced microbiota depletion prior to silica exposure reduced silica-mediated lung fibrosis and inflammation.
CONCLUSION
Lung microbiota is associated with silica-induced lung injury. Overproduction of SP-A and SP-D, induced by Abx-induced microbiota depletion, may enhance the resistance of mouse lung tissue to silica-induced injury.
Animals
;
Silicosis/prevention & control*
;
Lung/metabolism*
;
Mice
;
Anti-Bacterial Agents/pharmacology*
;
Microbiota/drug effects*
;
Silicon Dioxide/toxicity*
;
Mice, Inbred C57BL
;
Male
;
Pulmonary Surfactant-Associated Proteins/genetics*
6.Phenotypic Function of Legionella pneumophila Type I-F CRISPR-Cas.
Ting MO ; Hong Yu REN ; Xian Xian ZHANG ; Yun Wei LU ; Zhong Qiu TENG ; Xue ZHANG ; Lu Peng DAI ; Ling HOU ; Na ZHAO ; Jia HE ; Tian QIN
Biomedical and Environmental Sciences 2025;38(9):1105-1119
OBJECTIVE:
CRISPR-Cas protects bacteria from exogenous DNA invasion and is associated with bacterial biofilm formation and pathogenicity.
METHODS:
We analyzed the type I-F CRISPR-Cas system of Legionella pneumophila WX48, including Cas1, Cas2-Cas3, Csy1, Csy2, Csy3, and Cas6f, along with downstream CRISPR arrays. We explored the effects of the CRISPR-Cas system on the in vitro growth, biofilm-forming ability, and pathogenicity of L. pneumophila through constructing gene deletion mutants.
RESULTS:
The type I-F CRISPR-Cas system did not affect the in vitro growth of wild-type or mutant strains. The biofilm formation and intracellular proliferation of the mutant strains were weaker than those of the wild type owing to the regulation of type IV pili and Dot/Icm type IV secretion systems. In particular, Cas6f deletion strongly inhibited these processes.
CONCLUSION
The type I-F CRISPR-Cas system may reduce biofilm formation and intracellular proliferation in L. pneumophila.
Legionella pneumophila/pathogenicity*
;
CRISPR-Cas Systems
;
Biofilms/growth & development*
;
Phenotype
;
Bacterial Proteins/metabolism*
;
Gene Deletion
7.Engineering of CmpLs enhances L-glutamate production of Corynebacterium glutamicum.
Xingtao ZUO ; Shasha ZHONG ; Ningyun CAI ; Tuo SHI ; Zhidan ZHANG ; Yuantao LIU ; Jiao LIU ; Depei WANG ; Jiuzhou CHEN ; Ping ZHENG
Chinese Journal of Biotechnology 2025;41(1):271-287
The efficient production of L-glutamate is dependent on the product's rapid efflux, hence researchers have recently concentrated on artificially modifying its transport system and cell membrane wall structure. Considering the unique composition and structure of the cell wall of Corynebacterium glutamicum, we investigated the effects of CmpLs on L-glutamate synthesis and transport in SCgGC7, a constitutive L-glutamate efflux strain. First, the knockout strains of CmpLs were constructed, and it was confirmed that the deletion of CmpL1 and CmpL4 significantly improved the performance of L-glutamate producers. Next, temperature-sensitive L-glutamate fermentation with the CmpL1 and CmpL4 knockout strains were carried out in 5 L bioreactors, where the knockout strains showcased temperature-sensitive characteristics and enhanced capacities for L-glutamate production under high temperatures. Notably, the CmpL1 knockout strain outperformed the control strain in terms of L-glutamate production, showing production and yield increases of 69.2% and 55.3%, respectively. Finally, the intracellular and extracellular metabolites collected at the end of the fermentation process were analyzed. The modification of CmpLs greatly improved the L-glutamate excretion and metabolic flux for both L-glutamate production and transport. Additionally, the CmpL1 knockout strain showed decreased accumulation of downstream metabolites of L-glutamate and intermediate metabolites of tricarboxylic acid (TCA) cycle, which were consistent with its high L-glutamate biosynthesis capacity. In addition to offering an ideal target for improving the stability and performance of the industrial strains for L-glutamate production, the functional complementarity and redundancy of CmpLs provide a novel target and method for improving the transport of other metabolites by modification of the cell membrane and cell wall structures in C. glutamicum.
Corynebacterium glutamicum/genetics*
;
Glutamic Acid/biosynthesis*
;
Fermentation
;
Metabolic Engineering
;
Bacterial Proteins/metabolism*
;
Bioreactors/microbiology*
;
Gene Knockout Techniques
8.Mining, characterization, and expression of a fructan sucrase for efficient conversion of soybean oligosaccharides.
Bin WANG ; Jingru YING ; Yuanyuan CHEN ; Zemin FANG ; Yazhong XIAO ; Wei FANG ; Dongbang YAO
Chinese Journal of Biotechnology 2025;41(1):333-351
The high content of sucrose and raffinose reduces the prebiotic value of soybean oligosaccharides. Fructan sucrases can catalyze the conversion of sucrose and raffinose to high-value products such as fructooligosaccharides and melibiose. To obtain a fructan sucrase that can efficiently convert soybean oligosaccharides, we first mined the fructan sucrase gene from microorganisms in the coastal areas of Xisha Islands and Bohai Bay and then characterized the enzymatic and catalytic properties of the enzyme. Finally, recombinant extracellular expression of this gene was carried out in Bacillus subtilis. The results showed that a novel fructan sucrase, BhLS 39, was mined from Bacillus halotolerans. With sucrose and raffinose as substrates, BhLS 39 showed the optimal temperatures of 50 ℃ and 55 ℃, optimal pH 5.5 for both, and Kcat/Km ratio of 3.4 and 6.6 L/(mmol·s), respectively. When 400 g/L raffinose was used as the substrate, the melibiose conversion rate was 84.6% after 30 min treatment with 5 U BhLS 39. Furthermore, BhLS 39 catalyzed the conversion of sucrose to produce levan-type-fructooligosaccharide and levan. Then, the recombinant extracellular expression of BhLS 39 in B. subtilis was achieved. The co-expression of the intracellular chaperone DnaK and the extracellular chaperone PrsA increased the extracellular activity of the recombinant BhLS 39 by 5.2 folds to 17 U/mL compared with that of the control strain. BhLS 39 obtained in this study is conducive to improving the quality and economic benefits of soybean oligosaccharides. At the same time, the strategy used here to enhance the extracellular expression of BhLS 39 will also promote the efficient recombinant expression of other proteins in B. subtilis.
Oligosaccharides/metabolism*
;
Glycine max/metabolism*
;
Bacillus subtilis/metabolism*
;
Sucrase/biosynthesis*
;
Raffinose/metabolism*
;
Fructans/metabolism*
;
Sucrose/metabolism*
;
Bacillus/genetics*
;
Recombinant Proteins/biosynthesis*
;
Bacterial Proteins/biosynthesis*
9.Expression and enzymatic characterization of a chitosanase with tolerance to a wide range of pH from Bacillus atrophaeus.
Wenjuan DU ; Awagul TURSUN ; Zhiqin DONG ; Huijuan MA ; Zhenghai MA
Chinese Journal of Biotechnology 2025;41(1):352-362
To screen and identify a chitosanase with high stability, we cloned the chitosanase gene from Bacillus atrophaeus with a high protease yield from the barren saline-alkali soil and expressed this gene in Escherichia coli. The expressed chitosanase of B. atrophaeus (BA-CSN) was purified by nickel-affinity column chromatography. The properties including optimal temperature, optimal pH, substrate specificity, and kinetic parameters of BA-CSN were characterized. The results showed that BA-CSN had the molecular weight of 31.13 kDa, the optimal temperature of 55 ℃, the optimal pH 5.5, and good stability at temperatures below 45 ℃ and pH 4.0-9.0. BA-CSN also had good stability within 4 h of pH 3.0 and 10.0, be activated by K+, Na+, Mn2+, Ca2+, Mg2+, and Co2+, (especially by Mn2+), and be inhibited by Fe3+, Cu2+, and Ag+. BA-CSN showcased the highest relative activity in the hydrolysis of colloidal chitosan, and it had good hydrolysis ability for colloidal chitin. Under the optimal catalytic conditions, BA-CSN demonstrated the Michaelis constant Km and maximum reaction rate Vmax of 9.94 mg/mL and 26.624 μmoL/(mL·min), respectively, for colloidal chitosan. In short, BA-CSN has strong tolerance to acids and alkali, possessing broad industrial application prospects.
Bacillus/genetics*
;
Hydrogen-Ion Concentration
;
Escherichia coli/metabolism*
;
Glycoside Hydrolases/biosynthesis*
;
Substrate Specificity
;
Enzyme Stability
;
Chitosan/metabolism*
;
Temperature
;
Kinetics
;
Cloning, Molecular
;
Bacterial Proteins/biosynthesis*
;
Recombinant Proteins/genetics*
10.Construction and optimization of 1, 4-butanediamine biosensor based on transcriptional regulator PuuR.
Junjie LIU ; Minmin JIANG ; Tong SUN ; Xiangxiang SUN ; Yongcan ZHAO ; Mingxia GU ; Fuping LU ; Ming LI
Chinese Journal of Biotechnology 2025;41(1):437-447
Biosensors have become powerful tools for real-time monitoring of specific small molecules and precise control of gene expression in biological systems. High-throughput sensors for 1, 4-butanediamine biosynthesis can greatly improve the screening efficiency of high-yielding 1, 4-butanediamine strains. However, the strategies for adapting the characteristics of biosensors are still rarely studied, which limits the applicability of 1, 4-butanediamine biosensors. In this paper, we propose the development of a 1, 4-butanediamine biosensor based on the transcriptional regulator PuuR, whose homologous operator puuO is installed in the constitutive promoter PgapA of Escherichia coli to control the expression of the downstream superfolder green fluorescent protein (sfGFP) as the reporter protein. Finally, the biosensor showed a stable linear relationship between the GFP/OD600 value and the concentration of 1, 4-butanediamine when the concentration of 1, 4-butanediamine was 0-50 mmol/L. The promoters with different strengths in the E. coli genome were used to modify the 1, 4-butanediamine biosensor, and the functional properties of the PuuR-based 1, 4-butanediamine biosensor were explored and improved, which laid the groundwork for high-throughput screening of engineered strains highly producing 1, 4-butanediamine.
Biosensing Techniques/methods*
;
Escherichia coli/metabolism*
;
Promoter Regions, Genetic/genetics*
;
Green Fluorescent Proteins/metabolism*
;
Transcription Factors/genetics*
;
Escherichia coli Proteins/genetics*
;
Diamines/metabolism*
;
Gene Expression Regulation, Bacterial

Result Analysis
Print
Save
E-mail