1.Effects of acupoint catgut embedding on gut microbiota and fecal short-chain fatty acids in Parkinson's disease patients with constipation.
Xiaomei ZHANG ; Jie JIANG ; Wenying LI ; Juping CHEN ; Yin HUANG ; Wei REN ; Yafang SONG ; Jianhua SUN
Chinese Acupuncture & Moxibustion 2025;45(11):1533-1540
OBJECTIVE:
To observe the effects of acupoint catgut embedding (ACE) on gut microbiota and fecal short-chain fatty acids (SCFAs) levels in patients with Parkinson's disease (PD) with constipation.
METHODS:
A total of 80 PD patients with constipation were randomly divided into an observation group and a control group, 40 cases in each group. Additionally, 40 healthy individuals were recruited as a healthy control group. The control group received conventional Western medical treatment for PD combined with polyethylene glycol (PEG), once daily for eight weeks. The observation group received additional ACE treatment at bilateral Tianshu (ST25), Zusanli (ST36), and Shangjuxu (ST37), once every two weeks for eight weeks. The healthy control group received no intervention. The spontaneous bowel movements (SBMs) per week and patient assessment of constipation quality of life (PAC-QOL) scores were assessed at baseline and after treatment in the two groups. Fecal samples were collected at the end of treatment for the observation and the control groups and at baseline for the healthy control group. Gut microbiota composition and diversity were analyzed using 16S rRNA method, and SCFA levels were measured using high-performance liquid chromatography (HPLC).
RESULTS:
Compared before treatment, the observation group showed a significant increase in SBMs (P<0.01), and PAC-QOL scores including physical discomfort, psychosocial discomfort, worry and concern, and total score were significantly reduced (P<0.01) after treatment; the control group also showed a reduction in PAC-QOL total score after treatment (P<0.01). After treatment, the observation group had significantly more SBMs (P<0.01), and lower PAC-QOL physical discomfort, psychosocial discomfort, worry and concern scores, and total score (P<0.01), and higher PAC-QOL satisfaction score (P<0.01) than the control group. Compared with the healthy control group, the control group showed decreased Chao1 and Ace indices (P<0.01). Compared with the healthy control group, the relative abundance of Prevotella and Roseburia was increased (P<0.05), while that of Enterobacter and Ruminococcus torques (six species in total) was decreased (P<0.05) in the control group. Compared with the control group, the observation group had increased relative abundance of Dialister, Parabacteroides, and Ruminococcus torques (P<0.05), and decreased relative abundance of Prevotella and Eubacterium ruminantium (P<0.05). Compared with the healthy control group, the control group had increased fecal SCFA levels (P<0.05); compared with the control group, the observation group had reduced fecal SCFA levels (P<0.05). Compared with the healthy control group, acetic acid, propionic acid, and butyric acid levels were elevated in the control group (P<0.05); compared with the control group, acetic acid, propionic acid, and butyric acid levels were decreased in the observation group (P<0.05).
CONCLUSION
ACE could increase spontaneous bowel movements and improve the quality of life in PD patients with constipation, which may be related to the regulation of gut microbiota composition and SCFA levels.
Humans
;
Constipation/metabolism*
;
Male
;
Gastrointestinal Microbiome
;
Acupuncture Points
;
Female
;
Middle Aged
;
Parkinson Disease/complications*
;
Aged
;
Fatty Acids, Volatile/metabolism*
;
Catgut
;
Feces/microbiology*
;
Acupuncture Therapy
;
Quality of Life
;
Adult
2.Intestinal metabolites in colitis-associated carcinogenesis: Building a bridge between host and microbiome.
Yating FAN ; Yang LI ; Xiangshuai GU ; Na CHEN ; Ye CHEN ; Chao FANG ; Ziqiang WANG ; Yuan YIN ; Hongxin DENG ; Lei DAI
Chinese Medical Journal 2025;138(16):1961-1972
Microbial-derived metabolites are important mediators of host-microbial interactions. In recent years, the role of intestinal microbial metabolites in colorectal cancer has attracted considerable attention. These metabolites, which can be derived from bacterial metabolism of dietary substrates, modification of host molecules such as bile acids, or directly from bacteria, strongly influence the progression of colitis-associated cancer (CAC) by regulating inflammation and immune response. Here, we review how microbiome metabolites short-chain fatty acids (SCFAs), secondary bile acids, polyamines, microbial tryptophan metabolites, and polyphenols are involved in the tumorigenesis and development of CAC through inflammation and immunity. Given the heated debate on the metabolites of microbiota in maintaining gut homeostasis, serving as tumor molecular markers, and affecting the efficacy of immune checkpoint inhibitors in recent years, strategies for the prevention and treatment of CAC by targeting intestinal microbial metabolites are also discussed in this review.
Humans
;
Gastrointestinal Microbiome/physiology*
;
Animals
;
Carcinogenesis/metabolism*
;
Colitis-Associated Neoplasms/microbiology*
;
Fatty Acids, Volatile/metabolism*
;
Bile Acids and Salts/metabolism*
;
Colitis/microbiology*
3.CXCR3 counteracts cisplatin-induced muscle atrophy by regulating E3 ubiquitin ligases, myogenic factors, and fatty acid β-oxidation pathways.
Miao-Miao XU ; Xiao-Guang LIU ; Li-Ming LU ; Zhao-Wei LI
Acta Physiologica Sinica 2025;77(2):255-266
This study aims to explore the role and mechanism of CXC chemokine receptor 3 (CXCR3) in cisplatin-induced skeletal muscle atrophy. Wild-type mice were divided into two groups: cisplatin group and control group (treated by normal saline). The results showed that, compared to the control group, the expression levels of CXCR3 mRNA and protein were significantly up-regulated in the skeletal muscle of the cisplatin group, suggesting that CXCR3 may play an important role in the model of cisplatin-induced skeletal muscle atrophy. To further investigate its role and potential mechanisms, CXCR3 knockout mice and wild-type mice were treated with cisplatin to induce skeletal muscle atrophy. The results revealed that CXCR3 knockout not only failed to alleviate cisplatin-induced skeletal muscle atrophy, but also further reduced body weight, skeletal muscle mass, and muscle fiber cross-sectional area. Further analysis showed that, in the cisplatin-induced muscle atrophy model, CXCR3 knockout significantly up-regulated the expression levels of E3 ubiquitin ligases in skeletal muscle and down-regulated the expression levels of myogenic regulatory factors. To explore the molecular mechanism by which CXCR3 gene deletion exacerbated cisplatin-induced skeletal muscle atrophy, transcriptomic sequencing was performed on the atrophied skeletal muscles of wild-type and CXCR3 knockout mice. The results showed that, compared to wild-type mice, 14 genes were significantly up-regulated and 12 genes were significantly down-regulated in the skeletal muscle of CXCR3 knockout mice. Gene set enrichment analysis (GSEA) revealed a significant enrichment of genes related to fatty acid β-oxidation. Quantitative real-time PCR validation results were consistent with the transcriptomic sequencing results. These findings suggest that CXCR3 may counteract cisplatin-induced skeletal muscle atrophy by up-regulating E3 ubiquitin ligases, down-regulating myogenic regulatory factors, and enhancing the recruitment of fatty acid β-oxidation-related genes.
Animals
;
Cisplatin/adverse effects*
;
Muscular Atrophy/physiopathology*
;
Mice
;
Receptors, CXCR3/metabolism*
;
Ubiquitin-Protein Ligases/metabolism*
;
Mice, Knockout
;
Oxidation-Reduction
;
Fatty Acids/metabolism*
;
Muscle, Skeletal/metabolism*
;
Mice, Inbred C57BL
;
Male
4.Mechanism analysis of ω-3 polyunsaturated fatty acids in alleviating oxidative stress and promoting osteogenic differentiation of MC3T3-E1 cells through activating Nrf2/NQO1 pathway.
Jiahui HUANG ; Long CHEN ; Chen XU ; Haojie YU ; Shishuai ZHOU ; Jianzhong GUAN
Chinese Journal of Reparative and Reconstructive Surgery 2025;39(11):1459-1467
OBJECTIVE:
To explore the mechanism by which ω-3 polyunsaturated fatty acids (hereinafter referred to as "ω-3") exert antioxidant stress protection and promote osteogenic differentiation in MC3T3-E1 cells, and to reveal the relationship between ω-3 and the key antioxidant stress pathway involving nuclear factor E2-related factor 2 (Nrf2) and NAD (P) H quinone oxidoreductase 1 (NQO1) in MC3T3-E1 cells.
METHODS:
The optimal concentration of H 2O 2 (used to establish the oxidative stress model of MC3T3-E1 cells in vitro) and the optimal intervention concentrations of ω-3 were screened by cell counting kit 8. MC3T3-E1 cells were divided into blank control group, oxidative stress group (H 2O 2), low-dose ω-3 group (H 2O 2+low-dose ω-3), and high-dose ω-3 group (H 2O 2+high-dose ω-3). After osteoblastic differentiation for 7 or 14 days, the intracellular reactive oxygen species (ROS) level was measured by fluorescence staining and flow cytometry, and the mitochondrial morphological changes were observed by biological transmission electron microscope; the expression levels of Nrf2, NQO1, heme oxygenase 1 (HO-1), Mitofusin 1 (Mfn1), and Mfn2 were detected by Western blot to evaluate the cells' antioxidant stress capacity; the expression levels of Runt-related transcription factor 2 (RUNX2) and osteocalcin (OCN) were detected by immunofluorescence staining and Western blot; osteogenic potential of MC3T3-E1 cells was evaluated by alkaline phosphatase (ALP) staining and alizarin red staining.
RESULTS:
Compared with the oxidative stress group, the content of ROS in the low and high dose ω-3 groups significantly decreased, and the protein expressions of Nrf2, NQO1, and HO-1 significantly increased ( P<0.05). At the same time, the mitochondrial morphology of MC3T3-E1 cells improved, and the expressions of mitochondrial morphology-related proteins Mfn1 and Mfn2 significantly increased ( P<0.05). ALP staining and alizarin red staining showed that the low-dose and high-dose ω-3 groups showed stronger osteogenic ability, and the expressions of osteogenesis-related proteins RUNX2 and OCN significantly increased ( P<0.05). And the above results showed a dose-dependence in the two ω-3 treatment groups ( P<0.05).
CONCLUSION
ω-3 can enhance the antioxidant capacity of MC3T3-E1 cells under oxidative stress conditions and upregulate their osteogenic activity, possibly through the Nrf2/NQO1 signaling pathway.
Oxidative Stress/drug effects*
;
NF-E2-Related Factor 2/metabolism*
;
NAD(P)H Dehydrogenase (Quinone)/metabolism*
;
Animals
;
Mice
;
Osteogenesis/drug effects*
;
Cell Differentiation/drug effects*
;
Fatty Acids, Omega-3/pharmacology*
;
Signal Transduction/drug effects*
;
Osteoblasts/drug effects*
;
Reactive Oxygen Species/metabolism*
;
Cell Line
;
Hydrogen Peroxide/pharmacology*
;
Core Binding Factor Alpha 1 Subunit/metabolism*
;
Antioxidants/pharmacology*
;
Heme Oxygenase-1/metabolism*
5.Research progress on glycolipid metabolism of Sertoli cell in the development of spermatogenic cell.
Shuhao LI ; Liang KONG ; Jingyan LIANG ; Tan MA
Journal of Zhejiang University. Medical sciences 2025;54(2):257-265
Sertoli cells play an important role in the process of spermatogenesis, and the abnormalities in spermatogenesis are closely related to disruptions in glycolipid metabolism. The metabolic environment of Sertoli cells is hypoxic, with glycolysis and fatty acid β-oxidation being the primary metabolic pathways. In Sertoli cells, glycolysis produces lactate to provide energy for spermatogenic cells, while fatty acid β-oxidation generates ATP. Currently, the relationship between glycolipid metabolism in Sertoli cells and spermatogenic cell development, as well as the interplay between glucose and lipid metabolism remain unclear. Various hormones, including sex hormones, can affect glucose metabolism in Sertoli cells by endocrine regulation. The activation or inhibition of signaling pathways such as AMPK, mTOR, and Akt can alter the expression levels of glycolysis-related transporter genes and the synthesis of fatty acids, thereby affecting glycolipid metabolism in Sertoli cells. Some transcription factors such as PPARγ can regulate downstream fatty acid metabolism-related genes by directly binding to their response elements and promoting the oxidation of fatty acids in Sertoli cells. In this article we elaborate on the key factors influencing glycolipid metabolism in Sertoli cells and their interconnections, as well as their potential clinical implications, offering new insights for precisely targeted treatments of male infertility.
Sertoli Cells/cytology*
;
Male
;
Glycolipids/metabolism*
;
Spermatogenesis/physiology*
;
Humans
;
Lipid Metabolism
;
Animals
;
Fatty Acids/metabolism*
;
Signal Transduction
;
Glycolysis
6.Advances in pharmacological research for retinopathy of prematurity.
Yanxi XIE ; Suilian ZHENG ; Hui YANG
Journal of Zhejiang University. Medical sciences 2025;54(3):411-421
Retinopathy of prematurity (ROP) is a proliferative retinal vascular disease that threatens the vision of premature infants. Various novel drugs have demonstrated therapeutic potential for ROP by targeting signaling pathways associated with vascular endothelial growth factor (VEGF) [such as PI3K/AKT, hypoxia-inducible factor (HIF)-1α/VEGF], oxidative stress, tumor necrosis factor (TNF)-α, and Notch pathways. Propranolol, insulin-like growth factor-1, and celecoxib attenuate pathological neovascularization via the PI3K/Akt signaling pathway. Tripterine and melatonin inhibit retinal neovascularization by modulating the HIF-1α/VEGF signaling axis. Adiponectin mitigates the damage caused by oxidative stress and preserves endothelial function by enhancing endothelial nitric oxide synthase activity. Omega-3 polyunsaturated fatty acids suppress TNF-α-mediated inflammatory responses, modulate retinal development and angiogenesis, and reduce retinal neovascular lesions. DAPT, a γ-secretase inhibitor, blocks Notch signaling to suppress abnormal vascular proliferation. These agents exhibit synergistic multi-pathway anti-angiogenic effects in preclinical models and early-phase clinical trials, offering critical insights for advancing drug development and clinical translation in ROP management.
Retinopathy of Prematurity/metabolism*
;
Humans
;
Signal Transduction/drug effects*
;
Infant, Newborn
;
Vascular Endothelial Growth Factor A/metabolism*
;
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism*
;
Tumor Necrosis Factor-alpha/metabolism*
;
Oxidative Stress/drug effects*
;
Fatty Acids, Omega-3/therapeutic use*
;
Phosphatidylinositol 3-Kinases/metabolism*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Receptors, Notch/metabolism*
;
Angiogenesis Inhibitors/therapeutic use*
;
Insulin-Like Growth Factor I/therapeutic use*
7.The role of polyunsaturated fatty acid lipid peroxidation in ferroptosis after intracerebral hemorrhage: a review of mecha-nisms and therapeutic implications.
Man GUO ; Guohui ZHAO ; Zhibiao CAI ; Zhenyu ZHANG ; Jie ZHOU
Journal of Zhejiang University. Medical sciences 2025;54(5):694-704
Ferroptosis, a regulated cell death process distinct from apoptosis, is characterized by iron dysregulation and reactive oxygen species (ROS) accumulation. After intracerebral hemorrhage (ICH), decreased cerebral blood flow and iron released from erythrocytes trigger lipid peroxidation-particularly of polyunsaturated fatty acids (PUFAs)-through a cascade of reactions in local brain tissues, promoting ferroptosis. Mitochondrial dysfunction and neuroinflammation further elevate ROS, exacerbating lipid peroxidation and accelerating neuronal ferroptosis. Thus, PUFA peroxidation and associated metabolic pathways play a critical role in ICH-related neuronal damage. This review summarizes current understanding of how PUFA peroxidation contributes to ferro-ptosis after ICH, discusses key regulatory mechanisms involving lipid and iron metabolism, and highlights potential therapeutic strategies targeting ferroptosis to improve neurological outcomes.
Ferroptosis/physiology*
;
Humans
;
Cerebral Hemorrhage/pathology*
;
Lipid Peroxidation
;
Fatty Acids, Unsaturated/metabolism*
;
Reactive Oxygen Species/metabolism*
;
Iron/metabolism*
;
Animals
;
Mitochondria/metabolism*
8.Qishen Granules Modulate Metabolism Flexibility Against Myocardial Infarction via HIF-1 α-Dependent Mechanisms in Rats.
Xiao-Qian SUN ; Xuan LI ; Yan-Qin LI ; Xiang-Yu LU ; Xiang-Ning LIU ; Ling-Wen CUI ; Gang WANG ; Man ZHANG ; Chun LI ; Wei WANG
Chinese journal of integrative medicine 2025;31(3):215-227
OBJECTIVE:
To assess the cardioprotective effect and impact of Qishen Granules (QSG) on different ischemic areas of the myocardium in heart failure (HF) rats by evaluating its metabolic pattern, substrate utilization, and mechanistic modulation.
METHODS:
In vivo, echocardiography and histology were used to assess rat cardiac function; positron emission tomography was performed to assess the abundance of glucose metabolism in the ischemic border and remote areas of the heart; fatty acid metabolism and ATP production levels were assessed by hematologic and biochemical analyses. The above experiments evaluated the cardioprotective effect of QSG on left anterior descending ligation-induced HF in rats and the mode of energy metabolism modulation. In vitro, a hypoxia-induced H9C2 model was established, mitochondrial damage was evaluated by flow cytometry, and nuclear translocation of hypoxia-inducible factor-1 α (HIF-1 α) was observed by immunofluorescence to assess the mechanism of energy metabolism regulation by QSG in hypoxic and normoxia conditions.
RESULTS:
QSG regulated the pattern of glucose and fatty acid metabolism in the border and remote areas of the heart via the HIF-1 α pathway, and improved cardiac function in HF rats. Specifically, QSG promoted HIF-1 α expression and entry into the nucleus at high levels of hypoxia (P<0.05), thereby promoting increased compensatory glucose metabolism; while reducing nuclear accumulation of HIF-1 α at relatively low levels of hypoxia (P<0.05), promoting the increased lipid metabolism.
CONCLUSIONS
QSG regulates the protein stability of HIF-1 α, thereby coordinating energy supply balance between the ischemic border and remote areas of the myocardium. This alleviates the energy metabolism disorder caused by ischemic injury.
Animals
;
Myocardial Infarction/physiopathology*
;
Male
;
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism*
;
Rats, Sprague-Dawley
;
Glucose/metabolism*
;
Drugs, Chinese Herbal/therapeutic use*
;
Energy Metabolism/drug effects*
;
Rats
;
Fatty Acids/metabolism*
;
Myocardium/pathology*
9.Research progress in the role of gut microbiota in ethanol metabolism.
Yuchun YANG ; Xiaojie ZHANG ; Ti CHEN
Journal of Central South University(Medical Sciences) 2025;50(3):501-510
In recent years, gut microbiota has been increasingly recognized as a key player in ethanol metabolism and the development of related diseases. On one hand, ethanol intake directly affects the gut, leading to significant alterations in microbial diversity and composition. On the other hand, gut microbiota influences ethanol-induced damage to various organs, especially the liver, through multiple metabolic byproducts (such as short-chain fatty acids like butyrate, propionate, and acetate), modulation of immune responses, alteration of intestinal barrier function, and regulation of ethanol-metabolizing enzymes. Given the close association between gut microbiota and ethanol metabolism, the gut microbiome presents a promising therapeutic target for alcohol-related liver diseases. This review summarizes recent advances in understanding how gut microbiota affects ethanol metabolism, aiming to elucidate its role in the onset and progression of ethanol-related diseases and to provide a theoretical basis and novel targets for microbiota-based interventions.
Gastrointestinal Microbiome/physiology*
;
Ethanol/metabolism*
;
Humans
;
Fatty Acids, Volatile/metabolism*
;
Liver Diseases, Alcoholic/metabolism*
;
Animals
;
Alcohol Drinking/metabolism*
10.Effects of metformin on gut microbiota and short-/medium-chain fatty acids in high-fat diet rats.
Ying SHI ; Lin XING ; Shanyu WU ; Fangzhi YUE ; Tianqiong HE ; Jing ZHANG ; Lingxuan OUYANG ; Suisui GAO ; Dongmei ZHANG ; Zhijun ZHOU
Journal of Central South University(Medical Sciences) 2025;50(5):851-863
OBJECTIVES:
Recent evidence suggests that the gut may be a primary site of metformin action. However, studies on the effects of metformin on gut microbiota remain limited, and its impact on gut microbial metabolites such as short-/medium-chain fatty acids is unclear. This study aims to investigate the effects of metformin on gut microbiota, short-/medium-chain fatty acids, and associated metabolic benefits in high-fat diet rats.
METHODS:
Twenty-four Sprague-Dawley rats were randomly divided into 3 groups: 1) Normal diet group (ND group), fed standard chow; 2) high-fat diet group (HFD group), fed a high-fat diet; 3) high-fat diet + metformin treatment group (HFD+Met group), fed a high-fat diet for 8 weeks, followed by daily intragastric administration of metformin solution (150 mg/kg body weight) starting in week 9. At the end of the experiment, all rats were sacrificed, and serum, liver, and colonic contents were collected for assessment of glucose and lipid metabolism, liver pathology, gut microbiota composition, and the concentrations of short-/medium-chain fatty acids.
RESULTS:
Metformin significantly improved HFD-induced glucose and lipid metabolic disorders and liver injury. Compared with the HFD group, the HFD+Met group showed reduced abundance of Blautia, Romboutsia, Bilophila, and Bacteroides, while Lactobacillus abundance significantly increased (all P<0.05). Colonic contents of butyric acid, 2-methyl butyric acid, valeric acid, octanoic acid, and lauric acid were significantly elevated (all P<0.05), whereas acetic acid, isoheptanoic acid, and nonanoic acid levels were significantly decreased (all P<0.05). Spearman correlation analysis revealed that Lactobacillus abundance was negatively correlated with body weight gain and insulin resistance, while butyrate and valerate levels were negatively correlated with insulin resistance and liver injury (all P<0.05).
CONCLUSIONS
Metformin significantly increases the abundance of beneficial bacteria such as Lactobacillus and promotes the production of short-/medium-chain fatty acids including butyric, valeric, and lauric acid in the colonic contents of HFD rats, suggesting that metformin may regulate host metabolism through modulation of the gut microbiota.
Animals
;
Metformin/pharmacology*
;
Rats, Sprague-Dawley
;
Diet, High-Fat/adverse effects*
;
Rats
;
Gastrointestinal Microbiome/drug effects*
;
Male
;
Fatty Acids, Volatile/metabolism*
;
Fatty Acids/metabolism*

Result Analysis
Print
Save
E-mail