1.Antagonistic effect of Lactobacillus reuteri on testicular reproductive toxicity of neonicotinoid insecticides in mice.
Zhen-Han XU ; Pei-Gen CHEN ; Jin-Tao GUO ; Lin-Yan LÜ ; Hai-Cheng CHEN ; Gui-Hua LIU
National Journal of Andrology 2025;31(2):131-137
OBJECTIVE:
To explore the effect of Lactobacillus reuteri on testicular injury in mice exposed to neonicotinoid insecticides (NNI).
METHODS:
Fifteen C57BL/6 male mice were randomly divided into control group (CTRL group), exposure group (NNI group) and Lactobacillus intervention group (NNI-L group). The mice in CTRL group were given 0.02ml/g of 0.5% carboxymethyl cellulose sodium solution by gavage for 14 days. The mice in NNI group were given 0.02 ml/g of NNI mixture by gavage for 14 days. The mice in NNI-L group were given 0.02 ml/g of NNI mixture by gavage and 5×108cfu/ml of Lactobacillus reuteri powder solution for 14 days. Then, the histomorphology and function of testicle were evaluated by hematoxylin-eosin staining, immunofluorescence staining and RNA sequencing.
RESULTS:
Compared with CTRL group, the thickness of testicular seminiferous epithelium in the NNI group was significantly thinner. And the decline in the number of spermatogenic cells and sperm was observed. And the expression of spermatogonial stem cell marker UCHL1 was down-regulated which was significantly improved in NNI-L group compared with the NNI group. The abnormal expressions of hormone and sperm methylation related genes in testis of NNI group were detected by RNA sequencing, with significant down-regulation being found in NPFF and IGF2. While the expression of HSD3B8 was significantly up-regulated. The abnormal expression of these genes could be significantly improved after oral administration of Lactobacillus reuteri.
CONCLUSION
Testicular spermatogenesis and endocrine function can be damaged by NNI exposure. And oral administration of Lactobacillus reuteri protects testis from the adverse effects of NNI toxicity.
Animals
;
Male
;
Limosilactobacillus reuteri
;
Testis/pathology*
;
Mice
;
Mice, Inbred C57BL
;
Insecticides/toxicity*
;
Neonicotinoids/toxicity*
;
Probiotics
;
Spermatogenesis/drug effects*
2.Mechanisms by which the gut microbiota regulates depressive disorder via the tryptophan metabolic pathway.
Jing DU ; Jiao LI ; Pule LIU ; Yan ZHANG ; Qiangli DONG ; Ning YANG ; Xinru LIU
Journal of Central South University(Medical Sciences) 2025;50(7):1263-1270
The relationship between gut microbiota and depressive disorder has become a research focus in recent years. Within the microbiota-gut-brain axis, the gut microbiota influences the onset and progression of depressive disorder primarily through the tryptophan metabolic pathway. Tryptophan, an essential amino acid in humans, is subject to dual regulation by intestinal microorganisms, which modulate its metabolic balance via inflammatory stimulation and microbial metabolite production. In depression, excessive activation of the kynurenine branch of tryptophan metabolism leads to the accumulation of proinflammatory and neurotoxic metabolites, thereby exacerbating neuroinflammation in the brain. Intervention studies indicate that the antidepressant-like effects of probiotics and traditional Chinese medicine are associated with remodeling of the gut microbiota, restoration of tryptophan metabolic balance, and alleviation of neuroinflammation. Furthermore, targeted inhibition of kynurenine 3-monooxygenase can mitigate neuroinflammation by regulating microglial activity, thus improving depressive-like behaviors. In summary, the metabolite-inflammation axis represents a central node in the interaction regulation between tryptophan metabolism and the microbiota-gut-brain axis. This provides a theoretical foundation for developing novel therapeutic strategies targeting depression through modulation of gut microbiota-mediated tryptophan metabolism.
Tryptophan/metabolism*
;
Gastrointestinal Microbiome/physiology*
;
Humans
;
Depressive Disorder/microbiology*
;
Probiotics/therapeutic use*
;
Brain/metabolism*
;
Kynurenine/metabolism*
;
Metabolic Networks and Pathways
;
Animals
;
Medicine, Chinese Traditional
3.Early life Bifidobacterium bifidum BD-1 intervention alleviates hyperactivity of juvenile female rats with attention deficit hyperactivity disorder.
Yang YANG ; Kai WANG ; Jianxiu LIU ; Zhimo ZHOU ; Wen JIA ; Simou WU ; Jinxing LI ; Fang HE ; Ruyue CHENG
Journal of Southern Medical University 2025;45(4):702-710
OBJECTIVES:
To investigate the effects of early life intervention with Bifidobacterium bifidum BD-1 (B. bifidum BD-1) on hyperactivity in a female mouse model of attention deficit hyperactivity disorder (ADHD) and explore the underlying mechanisms.
METHODS:
Eight newborn female Wistar-Kyoto (WKY) rats and 6 spontaneous hypertensive rats (SHRs) were gavaged with saline and another 6 SHRs were gavaged with B. bifidum BD-1 (109 CFU) daily for 3 weeks. Open field test of the rats was conducted at 7 weeks, and fecal samples were collected at weaning (3 weeks) and at 7 weeks for 16S rRNA sequencing. Immunofluorescent staining was used to detect dopamine transporter (DAT) and tyrosine hydroxylase (Th) levels in the striatum and activated microglia in the prefrontal cortex. Treg cells in the mesenteric lymph nodes, spleen and blood were analyzed using flow cytometry.
RESULTS:
The SHRs traveled a significantly greater distance in open fields test than WKY rats, and this behavior was significantly attenuated by B. bifidum BD-1 intervention. The expression of DAT and Th in the striatum was significantly lower in the SHRs than in WKY rats, while B. bifidum BD-1 treatment obviously increased Th levels in the SHRs. B. bifidum BD-1 intervention significantly deceased the number of activated microglia and increased Treg cell counts in the spleen of SHRs. The treatment also enhanced α diversity in gut microbiota of the SHRs and resulted in a decreased Firmicutes/Bacteroidota ratio, more active Muribaculaceae growth, and suppression of Clostridia_UCG-014 proliferation.
CONCLUSIONS
Early life intervention with B. bifidum BD-1 alleviates hyperactivity in female SHRs by modulating the gut microbiota and peripheral immune response, suppressing neuroinflammation and improving dopaminergic system function. These findings provide evidence for early prevention strategies and support the development and application of psychobiotics for ADHD.
Animals
;
Female
;
Rats
;
Rats, Inbred WKY
;
Rats, Inbred SHR
;
Attention Deficit Disorder with Hyperactivity/therapy*
;
Bifidobacterium bifidum
;
Probiotics/therapeutic use*
;
Dopamine Plasma Membrane Transport Proteins/metabolism*
;
Tyrosine 3-Monooxygenase/metabolism*
;
Gastrointestinal Microbiome
;
Disease Models, Animal
4.Lactobacillus plantarum ZG03 alleviates oxidative stress via its metabolites short-chain fatty acids.
Shuxian LIN ; Lina GUO ; Yan MA ; Yao XIONG ; Yingxi HE ; Xinzhu XU ; Wen SHENG ; Suhua XU ; Feng QIU
Journal of Southern Medical University 2025;45(10):2223-2230
OBJECTIVES:
To investigate the efficacy of Lactobacillus plantarum ZG03 (L. plantarum ZG03) for ameliorating oxidative stress in zebrafish.
METHODS:
We evaluated the growth pattern of L. plantarum ZG03, observed its morphology using field emission scanning electron microscopy, and assessed its safety and potential efficacy with whole-genome sequencing for genetic analysis. FITC-labeled ZG03 was used to observe its intestinal colonization in zebrafish. In a zebrafish model of 2% glucose-induced oxidative stress, the effect of ZG03 was evaluated by assessing the changes in neutrophils in the caudal hematopoietic tissue (CHT), superoxide dismutase (SOD) activity, reactive oxygen species (ROS) levels, and malondialdehyde (MDA) content. Liquid chromatography-mass spectrometry-based targeted metabolomics was used for analyzing short-chain fatty acids (SCFAs) in the zebrafish, and the antioxidant effects of the key metabolites (acetate, propionate, and caproate) were tested.
RESULTS:
On MRS agar, L. plantarum ZG03 formed circular, smooth, moist, and milky-white colonies with a rod-shaped cell morphology. Genomic analysis revealed abundant sugar metabolism gene clusters. After inoculation of FITC-labeled L. plantarum ZG03 in zebrafish, green fluorescence was clearly observed in the intestinal bulb, mid-intestine, and hind intestine. In zebrafish with glucose-induced oxidative stress, L. plantarum ZG03 significantly reduced ROS levels and the number of neutrophils in the CHT with increased SOD activity. L.plantarum ZG03 significantly increased the content of SCFAs including acetic acid, propionic acid, and caproic acid in zebrafish metabolites. In addition, sodium acetate, sodium propionate, and sodium caproate in the SCFAs significantly increased SOD activity in the zebrafish models.
CONCLUSIONS
L. plantarum ZG03 ameliorates oxidative stress in a glucose-induced zebrafish model through its metabolites, particularly the SCFAs including acetic acid, propionic acid and caproic acid.
Animals
;
Zebrafish/metabolism*
;
Oxidative Stress
;
Lactobacillus plantarum/metabolism*
;
Fatty Acids, Volatile/metabolism*
;
Probiotics
;
Reactive Oxygen Species/metabolism*
;
Superoxide Dismutase/metabolism*
5.Gut microbiota: new perspective on the treatment of acute pancreatitis and clinical application prospects.
Qun LANG ; Yujie ZENG ; Hua YAO ; Ninan DAI ; Xiaoyun FU ; Bao FU
Chinese Critical Care Medicine 2025;37(9):797-801
Acute pancreatitis (AP) is a severe inflammatory disease characterized by self-digestion of pancreatic tissue and inflammatory responses. Recent studies have revealed a close connection between gut microbiota and AP. The gut microbiota community, a complex ecosystem composed of trillions of microorganisms, is closely associated with various physiological activities of the host, including metabolic processes, immune system regulation, and intestinal structure maintenance. However, in patients with AP, dysbiosis of the gut microbiota are believed to play a key role in the occurrence and progression of the disease. This dysbiosis not only impairs the integrity of the intestinal barrier, but may also exacerbate inflammatory responses through multiple mechanisms, thereby affecting the severity of the disease and patient' clinical prognosis. This article reviews the mechanisms of action of gut microbiota in AP, explores how gut microbiota dysbiosis affects disease progression, and evaluates current clinical treatment methods to regulate intestinal flora, including probiotic supplementation, fecal microbiota transplantation, antibiotic therapy, and early enteral nutrition. In addition, this article discusses the efficacy and safety of the aforementioned therapeutic approaches, and outlines future research directions, aiming to provide novel perspectives and strategies for the diagnosis, treatment and prognostic evaluation of AP. Through in-depth understanding the interaction between gut microbiota and AP, it is expected that more precise and personalized therapeutic regimens will be developed to improve patients' quality of life and clinical outcomes.
Humans
;
Gastrointestinal Microbiome
;
Dysbiosis
;
Pancreatitis/microbiology*
;
Fecal Microbiota Transplantation
;
Probiotics/therapeutic use*
;
Acute Disease
;
Anti-Bacterial Agents/therapeutic use*
;
Enteral Nutrition
6.Preparation, optimization, and in vitro evaluation of Pediococcus acidilactici HRQ-1 microcapsules.
Ruiqin HAN ; Song XU ; Xinyuan WANG ; Jingjing WANG ; Xiaoxia ZHANG ; Liping DU ; Zhiyong HUANG
Chinese Journal of Biotechnology 2025;41(4):1415-1427
We have isolated an intestinal probiotic strain, Pediococcus acidilactici HRQ-1. To improve its gastrointestinal fluid tolerance, transportation and storage stability, and slow-release properties, we employed the extrusion method to prepare the microcapsules with P. acidilactici HRQ-1 as the core material and sodium alginate and chitosan as the wall material. The optimal conditions for preparing the microcapsules were determined by single factor and orthogonal tests, and the optimal ratio was determined by taking the embedding rate, survival rate, storage stability, gastrointestinal fluid tolerance, and release rate as the evaluation indexes. The results showed that under the optimal embedding conditions, the embedding rate reached (89.60±0.02)%. Under the optimal formula of freeze-drying protective agent, the freeze-drying survival rate reached (76.42±0.13)%, and the average size of the microcapsules produced was (1.16±0.03) mm. The continuous gastrointestinal fluid simulation experiments confirmed that the microcapsules ensured the viable bacterial count and can slowly release bacteria in the intestinal fluid. The curve of the viable bacterial count during storage at 4 ℃ and room temperature indicated that the prepared microcapsules achieved strains' live number protection. The formula and preparation process of P. acidilactici microcapsules may provide a technological reserve for the preparation of more live bacterial drugs in the future.
Pediococcus acidilactici/chemistry*
;
Probiotics/chemistry*
;
Capsules/chemistry*
;
Alginates/chemistry*
;
Chitosan/chemistry*
;
Drug Compounding/methods*
;
Glucuronic Acid/chemistry*
;
Hexuronic Acids/chemistry*
;
Freeze Drying
7.Association between gut microbiota and hyperuricemia: insights into innovative therapeutic strategies.
Shujuan ZHANG ; Xiaoqiu LIU ; Yuxin ZHONG ; Yu FU
Chinese Journal of Biotechnology 2025;41(6):2290-2309
Uric acid (UA) is the final metabolite of purines in the human body. An imbalance in UA production and excretion that disrupts homeostasis leads to elevated blood UA levels and the development of hyperuricemia (HUA). Approximately one-third of UA is excreted through the intestinal tract. As a crucial component of the intestinal microenvironment, the gut microbiota plays a pivotal role in regulating blood UA levels. Alterations or imbalances in gut microbiota composition are linked to the onset of HUA, which implies the potential of gut microbiota as a novel target for the prevention and treatment of HUA. This review introduces the occurrence mechanism and damage of hyperuricemia, examines the association between HUA and the gut microbiota and their metabolites, and explores the molecular mechanisms underlying gut microbiota-targeted therapies for HUA. Furthermore, it discusses the potential applications of probiotics, prebiotics, and traditional Chinese medicine (including both single herbs and compound formulas) with UA-lowering effects, along with cutting-edge technologies such as fecal microbiota transplantation and machine learning in HUA treatment. This review provides valuable perspectives and strategies for improving the prevention and treatment of HUA.
Hyperuricemia/microbiology*
;
Humans
;
Gastrointestinal Microbiome/physiology*
;
Probiotics/therapeutic use*
;
Uric Acid/blood*
;
Fecal Microbiota Transplantation
;
Prebiotics
;
Medicine, Chinese Traditional
8.Construction of an engineered probiotic strain for efficiently delivering chemokine CXCL12 and application of the strain in diabetic chronic wound healing.
Shengjie LI ; Huijuan SU ; Xiaoting LI ; Jing WEI ; Tingtao CHEN
Chinese Journal of Biotechnology 2025;41(6):2334-2348
Diabetic chronic wounds are characterized by difficult healing, recurrent progression, and high rates of disability and mortality, which make their clinical treatment a medical challenge urgent to be addressed. However, the complex local microenvironment conditions of chronic wounds, such as high protease activity and persistent inflammatory responses, result in low bioavailability of exogenous cytokines (e.g., chemokine CXCL12) at the wound site, limiting their clinical application. In this study, we utilized Lactobacillus plantarum WCFS1 as the chassis to develop an efficient CXCL12 delivery system based on synthetic biology. Subsequently, we evaluated the role of the engineered probiotic strain in promoting the chronic wound healing in diabetic mice. Firstly, we fused the endogenous secretion signal peptide lp_3050 (SPlp_3050) of L. plantarum WCFS1 and the commonly used secretion signal peptide usp45 (SPusp45) of lactic acid bacteria with the reporter gene gusA and inserted them into the pTRK892-P32(pgm) plasmid by molecular cloning. Then, we prepared the engineered strains and characterized the efficacy of the two signal peptides in driving the secretion of GusA. The results showed that SPlp_3050 efficiently drove the secretion of GusA in L. plantarum WCFS1, increasing the activity of GusA in the culture supernatant by nearly five times compared with that of SPlp_3050. Further, we fused SPlp_3050 and codon-optimized CXCL12 gene to construct an engineered probiotic strain Lpw-CXCL12 for CXCL12 delivery. The results demonstrated that the content of CXCL12 in the culture supernatant reached (13.40±0.20) μg/mL. Finally, we found that the engineered probiotic strain Lpw-CXCL12 accelerated chronic wound healing in a diabetic mouse model. In conclusion, these results support an engineered probiotic strain in promoting diabetic chronic wound healing, providing a new strategy and technological foundation for the management of diabetic chronic wounds in the future.
Probiotics
;
Animals
;
Chemokine CXCL12/biosynthesis*
;
Mice
;
Wound Healing
;
Lactobacillus plantarum/metabolism*
;
Diabetes Mellitus, Experimental/complications*
;
Male
9.Characteristics of gut microbiota determine effects of specific probiotics strains in patients with functional constipation.
Haohao ZHANG ; Lijuan SUN ; Zhixin ZHAO ; Yao ZHOU ; Yuyao LIU ; Nannan ZHANG ; Junya YAN ; Shibo WANG ; Renlong LI ; Jing ZHANG ; Xueying WANG ; Wenjiao LI ; Yan PAN ; Meixia WANG ; Bing LUO ; Mengbin LI ; Zhihong SUN ; Yongxiang ZHAO ; Yongzhan NIE
Chinese Medical Journal 2024;137(1):120-122
10.The efficacy and safety of oral probiotics for acne vulgaris: A systematic review and meta analysis of randomized controlled trials
Maria Leanna S. Caylao ; Benedicto Dl Carpio ; Eileen R. Morales ; Armelia L. Torres ; Faye Elinore V. Kison ; Matthew David S. Parco
Journal of the Philippine Dermatological Society 2024;33(Suppl 1):38-38
BACKGROUND
Long-term administration of conventional acne medications implies the potential risk of increasing microbial resistance, and safety risks. Recent interest has grown in probiotics as a potential treatment and as an adjunct for acne vulgaris.
OBJECTIVETo conduct a systematic review of randomized controlled trials of oral probiotics in reducing the severity of acne vulgaris compared to placebo or conventional treatments.
METHODSPubMed, Cochrane Central Register of Controlled Trials, Embase, WHO ICTRP and ClinicalTrials.gov were searched for relevant studies, and randomized controlled trials of acne vulgaris treatment with probiotic therapy were included. Outcomes include decrease in total lesion count/percent change from baseline, improvement of severity scores, inflammation related indicators, and gene expression. Changes in outcomes were calculated by effect size or pooled odds ratio.
RESULTSA total of 2 RCTs involving 100 participants were included. Both studies show low risk on the risk bias criteria. The data of 2 RCTs reporting severity scores (AGSS and GAGS) supports that probiotics can improve their scores and thereby their condition. For inflammation-related indicators, only 1 RCT showed that probiotics can improve the IGF1 and FOXO1 gene expression in the skin. Regarding safety, only Eguren et al. reported adverse event. Their RCT showed that the incidence of adverse events was low and no serious adverse events.
CONCLUSIONIn conclusion, the probiotics investigated in both studies may be given as adjuvant treatment for acne vulgaris as they improve the clinical course of patients.
Human ; Acne Vulgaris ; Probiotics ; Meta-analysis


Result Analysis
Print
Save
E-mail