1.Effect of Dictamni Cortex on Intestinal Barrier Damage by Untargeted Metabolomics and Targeted Metabolomics for Short-chain Fatty Acids
Xiaomin XU ; Donghua YU ; Yu WANG ; Pingping CHEN ; Jiameixue WO ; Suxia JIA ; Wenkai HU ; Fang LU ; Shumin LIU
Chinese Journal of Experimental Traditional Medical Formulae 2025;31(20):40-47
		                        		
		                        			
		                        			ObjectiveThis study aims to investigate the effect of Dictamni Cortex on intestinal barrier damage in rats and its mechanism by untargeted metabolomics and targeted metabolomics for short-chain fatty acids (SCFAs). MethodsRats were randomly divided into a control group, a high-dose group of Dictamni Cortex (8.1 g·kg-1), a medium-dose group (2.7 g·kg-1), and a low-dose group (0.9 g·kg-1). Except for the control group, the other groups were administered different doses of Dictamni Cortex by gavage for eight consecutive weeks. Hematoxylin-eosin (HE) staining was used to observe the pathological changes in the ileal tissue. Enzyme-linked immunosorbent assay (ELISA) was employed to detect the level of cytokines, including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β), in the ileal tissue of rats. Quantitative real-time fluorescence polymerase chain reaction (Real-time PCR) technology was used to detect the expression level of tight junction proteins, including zonula occludens-1 (ZO-1), Occludin, and Claudin-1 mRNAs, in the ileal tissue of rats to preliminarily explore the effects of Dictamni Cortex on intestinal damage. The dose with the most significant toxic phenotype was selected to further reveal the effects of Dictamni Cortex on the metabolic profile of ileal tissue in rats by non-targeted metabolomics combined with targeted metabolomics for SCFAs. ResultsCompared with the control group, all doses of Dictamni Cortex induced varying degrees of pathological damage in the ileum, increased TNF-α (P<0.01), IL-6 (P<0.01), and IL-1β (P<0.01) levels in the ileal tissue, and decreased the expression level of ZO-1 (P<0.05, P<0.01), Occludin (P<0.01), and Claudin-1 (P<0.05) in the ileal tissue, with the high-dose group showing the most significant toxic phenotypes. The damage mechanisms of the high-dose group of Dictamni Cortex on the ileal tissue were further explored by integrating non-targeted metabolomics and targeted metabolomics for SCFAs. The non-targeted metabolomics results showed that 21 differential metabolites were identified in the control group and the high-dose group. Compared with that in the control group, after Dictamni Cortex intervention, the level of 14 metabolites was significantly increased (P<0.05, P<0.01), and the level of seven metabolites was significantly decreased (P<0.05, P<0.01) in the ileal contents. These metabolites collectively acted on 10 related metabolic pathways, including glycerophospholipids and primary bile acid biosynthesis. The quantitative data of targeted metabolomics for SCFAs showed that Dictamni Cortex intervention disrupted the level of propionic acid, butyric acid, acetic acid, caproic acid, isobutyric acid, isovaleric acid, valeric acid, and isocaproic acid in the ileal contents of rats. Compared with those in the control group, the level of isobutyric acid, isovaleric acid, and valeric acid were significantly increased, while the level of propionic acid, butyric acid, and acetic acid were significantly decreased in the ileal contents of rats after Dictamni Cortex intervention (P<0.05, P<0.01). ConclusionDictamni Cortex can induce intestinal damage by regulating glycerophospholipid metabolism, primary bile acid biosynthesis, and metabolic pathways for SCFAs. 
		                        		
		                        		
		                        		
		                        	
2.Metabolomics Reveals Immune System Domage of Dictamnine
Xiaocan GAI ; Jiaxin RUAN ; Sujie LIU ; Chen WANG ; Xiaofan WANG ; Jiahe YAN ; Yu WANG ; Fang LU ; Shumin LIU
Chinese Journal of Experimental Traditional Medical Formulae 2025;31(20):57-65
		                        		
		                        			
		                        			ObjectiveTo explore the mechanism of the immunotoxicity induced by dictamnine (DIC) in rats and the recovery effect after drug withdrawal by ultra-performance liquid chromatography-quadrupole-time-of-flight mass spectrometry, thereby providing a theoretical basis for elucidating the toxic mechanism of DIC. MethodsSD rats were randomized into blank (normal saline), DIC (10 mg·kg-1), and DIC withdrawal (recovery period) groups (n=8). The rats were continuously treated for 7 days, once a day, and the body weight and organ weight were recorded. The levels of interleukin-1 (IL-1), IL-6, and tumor necrosis factor-α (TNF-α) in the serum and immunoglobulin A (IgA), immunoglobulin G (IgG), and immunoglobulin M (IgM) in the spleen were determined by enzyme-linked immunosorbent assay. Hematoxylin-eosin staining was used to observe the pathological changes in the spleen. ultra performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) was employed to screen the potential biomarkers of immune inflammation caused by DIC, and pathway enrichment analysis and correlation analysis were performed. The mRNA levels of IL-1β, TNF-α, lysophosphatidylcholine acyltransferase 2 (LPCAT2), and farnesoid X receptor (FXR) in the serum were determined by Real-time fluorescence quantitative polymerase chain reaction (Real-time PCR). ResultsCompared with the blank group, the DIC group showed elevated levels of IL-1β, IL-6, and TNF-α in the serum (P<0.01), and the DIC withdrawal group showcased lowered levels of IL-1β, IL-6, and TNF-α in the serum (P<0.01). The levels of IgA, IgG, and IgM in the spleen of rats in the DIC group were decreased (P<0.01), while those in the DIC withdrawal group were recovered (P<0.05, P<0.01). Untargeted metabolomics of the serum and spleen screened out 14 common differential metabolites and 14 common metabolic pathways. The Spearman correlation analysis between differential metabolites and inflammatory factors identified PC (32∶0), LysoPC (20∶4/0∶0), LysoPC (P-18∶0/0∶0), taurochenodeoxycholic acid, taurocholic acid, LysoPC [20∶5(5Z,8Z,11Z,14Z,17Z)/0∶0], chenodeoxycholic acid, arachidonic acid, LysoPC (18∶0/0∶0), LysoPC (15∶0/0∶0), LysoPC (16∶0/0∶0), and LysoPC (17∶0/0∶0) as the biomarkers of immunotoxicity induced by DIC in SD rats. In the process of immunotoxicity caused by DIC, lipid metabolism disorders such as glycerophospholipid metabolism, primary bile acid metabolism, and arachidonic acid metabolism were enriched, which was consistent with the DIC-induced inflammatory factors and pathological characteristics of the spleen. Compared with the blank group, the DIC group exhibited up-regulated mRNA levels of IL-1β, TNF-α, LPCAT2, and FXR (P<0.01), and the up-regulation was decreased in the withdrawal group (P<0.01). ConclusionDIC can lead to immune and inflammatory disorders. DIC withdrawal can regulate the expression of biomarkers related to serum and spleen metabolites, regulate the inflammatory metabolic pathway, reduce the inflammation level, and alleviate the metabolic disorders, thus attenuating the potential toxicity induced by DIC. 
		                        		
		                        		
		                        		
		                        	
3.Clinical application and research progress of artificial intelligence-assisted diagnosis of pulmonary nodules
Chen LIU ; Zemin FANG ; Zuoliang SHAO ; Ruoting YU ; Wei GAO
Chinese Journal of Clinical Thoracic and Cardiovascular Surgery 2025;32(06):846-854
		                        		
		                        			
		                        			Artificial intelligence (AI) has been widely used in all walks of life, including healthcare, and has shown great application value in the auxiliary diagnosis of pulmonary nodules in the medical field. In the face of a large amount of lung imaging data, clinicians use AI tools to identify lesions more quickly and accurately, improving work efficiency, but there are still many problems in this field, such as the high false positive rate of recognition, and the difficulty in identifying special types of nodules. Researchers and clinicians are actively developing and using AI tools to promote their continuous evolution and make them better serve human health. This article reviews the clinical application and research progress of AI-assisted diagnosis of pulmonary nodules.
		                        		
		                        		
		                        		
		                        	
4.Underlying Mechanism of Wuwei Shenqintang in Amelioration of Pulmonary Fibrosis by Regulating "Lung-intestine Axis" Based on UPLC-Q-TOF-MS Metabolomics Technology
Mengdi SUN ; Fang LU ; Donghua YU ; Yu WANG ; Pingping CHEN ; Shumin LIU
Chinese Journal of Experimental Traditional Medical Formulae 2025;31(21):11-20
		                        		
		                        			
		                        			ObjectiveTo explore the mechanism of action of Wuwei Shenqintang in improving pulmonary fibrosis by using ultra-performance liquid chromatography-quadrupole-time-of-flight mass spectrometry (UPLC-Q-TOF-MS) for metabolomic analysis of lung tissue and feces. MethodsA rat model with pulmonary fibrosis was established by intratracheal injection of 5 mg·kg-1 bleomycin. The successfully modeled rats were randomly divided into a blank group, a model group, a prednisone (3.15 mg·kg-1) group, and low-dose, medium-dose, and high-dose groups of Wuwei Shenqintang (4.586, 9.172, 18.344 g·kg-1). The rats were given intragastric administration once a day for 28 consecutive days. Hematoxylin-eosin (HE) staining was used to measure the pathological changes in lung and colon tissue, and Masson staining was used to detect the degree of pulmonary fibrosis. Enzyme-linked immunosorbent assay (ELISA) was used to detect the expression of interleukin-1β (IL-1β), IL-6, IL-8, tumor necrosis factor-α (TNF-α), and secretory immunoglobulin A (SIgA) in bronchoalveolar lavage fluid and intestinal mucus. Immunohistochemistry and reverse transcription quantitative polymerase chain reaction (Real-time PCR) were used to detect the expression of type Ⅰ collagen (Col-Ⅰ), fibronectin (FN), and alpha smooth muscle actin (α-SMA) in lung tissue. UPLC-Q-TOF-MS was used to study the changes in the metabolic network of lung tissue and feces in rats with pulmonary fibrosis treated with Wuwei Shenqintang, screen potential biomarkers for the treatment of pulmonary fibrosis by Wuwei Shenqintang, and perform pathway enrichment analysis. ResultsCompared with the blank group, the model group showed extensive inflammatory cell infiltration and continuous fibrotic lesions in lung tissue, colonic mucosal damage, and connective tissue hyperplasia. The expression of IL-6, IL-8, IL-1β, TNF-α, and SIgA in bronchoalveolar lavage fluid and intestinal mucus was significantly increased (P<0.01). The expression of Col-Ⅰ, FN, and α-SMA proteins and mRNAs in lung tissue was significantly upregulated (P<0.01). Compared with the model group, the groups of Wuwei Shenqintang exhibited significantly reduced inflammatory infiltration and blue collagen deposition in lung tissue, alleviated colonic damage, decreased expression of IL-6, IL-8, IL-1β, TNF-α, and SIgA in bronchoalveolar lavage fluid and intestinal mucus (P<0.01), and reduced average absorbance values and mRNA expression of Col-Ⅰ, FN, and α-SMA in lung tissue (P<0.05, P<0.01), with the prednisone group and the medium-dose and high-dose groups of Wuwei Shenqintang showing the most significant effects. The metabolomics results for lung tissue showed that compared with the blank group, the model group had 19 significantly different compounds (P<0.05, P<0.01). Wuwei Shenqintang could normalize 17 of these compounds compared with the model group (P<0.05, P<0.01). Fecal metabolomics results showed that compared with those in the blank group, there were 42 compounds with significant differences in the model group (P<0.05, P<0.01). Compared with the model control group, Wuwei Shenqintang could normalize 41 of these compounds (P<0.05, P<0.01). The combined analysis results indicated that Wuwei Shenqintang might inhibit pulmonary fibrosis by regulating the biosynthesis of phenylalanine, tyrosine, and tryptophan as well as the retinol metabolism pathway. ConclusionWuwei Shenqintang can ameliorate pulmonary fibrosis, which may be related to the regulation of the "lung-intestine axis". 
		                        		
		                        		
		                        		
		                        	
5.Underlying Mechanism of Wuwei Shenqintang in Amelioration of Pulmonary Fibrosis by Regulating "Lung-intestine Axis" Based on UPLC-Q-TOF-MS Metabolomics Technology
Mengdi SUN ; Fang LU ; Donghua YU ; Yu WANG ; Pingping CHEN ; Shumin LIU
Chinese Journal of Experimental Traditional Medical Formulae 2025;31(21):11-20
		                        		
		                        			
		                        			ObjectiveTo explore the mechanism of action of Wuwei Shenqintang in improving pulmonary fibrosis by using ultra-performance liquid chromatography-quadrupole-time-of-flight mass spectrometry (UPLC-Q-TOF-MS) for metabolomic analysis of lung tissue and feces. MethodsA rat model with pulmonary fibrosis was established by intratracheal injection of 5 mg·kg-1 bleomycin. The successfully modeled rats were randomly divided into a blank group, a model group, a prednisone (3.15 mg·kg-1) group, and low-dose, medium-dose, and high-dose groups of Wuwei Shenqintang (4.586, 9.172, 18.344 g·kg-1). The rats were given intragastric administration once a day for 28 consecutive days. Hematoxylin-eosin (HE) staining was used to measure the pathological changes in lung and colon tissue, and Masson staining was used to detect the degree of pulmonary fibrosis. Enzyme-linked immunosorbent assay (ELISA) was used to detect the expression of interleukin-1β (IL-1β), IL-6, IL-8, tumor necrosis factor-α (TNF-α), and secretory immunoglobulin A (SIgA) in bronchoalveolar lavage fluid and intestinal mucus. Immunohistochemistry and reverse transcription quantitative polymerase chain reaction (Real-time PCR) were used to detect the expression of type Ⅰ collagen (Col-Ⅰ), fibronectin (FN), and alpha smooth muscle actin (α-SMA) in lung tissue. UPLC-Q-TOF-MS was used to study the changes in the metabolic network of lung tissue and feces in rats with pulmonary fibrosis treated with Wuwei Shenqintang, screen potential biomarkers for the treatment of pulmonary fibrosis by Wuwei Shenqintang, and perform pathway enrichment analysis. ResultsCompared with the blank group, the model group showed extensive inflammatory cell infiltration and continuous fibrotic lesions in lung tissue, colonic mucosal damage, and connective tissue hyperplasia. The expression of IL-6, IL-8, IL-1β, TNF-α, and SIgA in bronchoalveolar lavage fluid and intestinal mucus was significantly increased (P<0.01). The expression of Col-Ⅰ, FN, and α-SMA proteins and mRNAs in lung tissue was significantly upregulated (P<0.01). Compared with the model group, the groups of Wuwei Shenqintang exhibited significantly reduced inflammatory infiltration and blue collagen deposition in lung tissue, alleviated colonic damage, decreased expression of IL-6, IL-8, IL-1β, TNF-α, and SIgA in bronchoalveolar lavage fluid and intestinal mucus (P<0.01), and reduced average absorbance values and mRNA expression of Col-Ⅰ, FN, and α-SMA in lung tissue (P<0.05, P<0.01), with the prednisone group and the medium-dose and high-dose groups of Wuwei Shenqintang showing the most significant effects. The metabolomics results for lung tissue showed that compared with the blank group, the model group had 19 significantly different compounds (P<0.05, P<0.01). Wuwei Shenqintang could normalize 17 of these compounds compared with the model group (P<0.05, P<0.01). Fecal metabolomics results showed that compared with those in the blank group, there were 42 compounds with significant differences in the model group (P<0.05, P<0.01). Compared with the model control group, Wuwei Shenqintang could normalize 41 of these compounds (P<0.05, P<0.01). The combined analysis results indicated that Wuwei Shenqintang might inhibit pulmonary fibrosis by regulating the biosynthesis of phenylalanine, tyrosine, and tryptophan as well as the retinol metabolism pathway. ConclusionWuwei Shenqintang can ameliorate pulmonary fibrosis, which may be related to the regulation of the "lung-intestine axis". 
		                        		
		                        		
		                        		
		                        	
6.Gradient artificial bone repair scaffold regulates skeletal system tissue repair and regeneration
Yu ZHANG ; Ruian XU ; Lei FANG ; Longfei LI ; Shuyan LIU ; Lingxue DING ; Yuexi WANG ; Ziyan GUO ; Feng TIAN ; Jiajia XUE
Chinese Journal of Tissue Engineering Research 2025;29(4):846-855
		                        		
		                        			
		                        			BACKGROUND:Gradient artificial bone repair scaffolds can mimic unique anatomical features in musculoskeletal tissues,showing great potential for repairing injured musculoskeletal tissues. OBJECTIVE:To review the latest research advances in gradient artificial bone repair scaffolds for tissue engineering in the musculoskeletal system and describe their advantages and fabrication strategies. METHODS:The first author of the article searched the Web of Science and PubMed databases for articles published from 2000 to 2023 with search terms"gradient,bone regeneration,scaffold".Finally,76 papers were analyzed and summarized after the screening. RESULTS AND CONCLUSION:(1)As an important means of efficient and high-quality repair of skeletal system tissues,gradient artificial bone repair scaffolds are currently designed bionically for the natural gradient characteristics of bone tissue,bone-cartilage,and tendon-bone tissue.These scaffolds can mimic the extracellular matrix of native tissues to a certain extent in terms of structure and composition,thus promoting cell adhesion,migration,proliferation,differentiation,and regenerative recovery of damaged tissues to their native state.(2)Advanced manufacturing technology provides more possibilities for gradient artificial bone repair scaffold preparation:Gradient electrospun fiber scaffolds constructed by spatially differentiated fiber arrangement and loading of biologically active substances have been developed;gradient 3D printed scaffolds fabricated by layered stacking,graded porosity,and bio-3D printing technology;gradient hydrogel scaffolds fabricated by in-situ layered injections,simple layer-by-layer stacking,and freeze-drying method;and in addition,there are also scaffolds made by other modalities or multi-method coupling.These scaffolds have demonstrated good biocompatibility in vitro experiments,were able to accelerate tissue regeneration in small animal tests,and were observed to have significantly improved histological structure.(3)The currently developed gradient artificial bone repair scaffolds have problems such as mismatch of gradient scales,unclear material-tissue interactions,and side effects caused by degradation products,which need to be further optimized by combining the strengths of related disciplines and clinical needs in the future.
		                        		
		                        		
		                        		
		                        	
7.Effect Analysis of Different Interventions to Improve Neuroinflammation in The Treatment of Alzheimer’s Disease
Jiang-Hui SHAN ; Chao-Yang CHU ; Shi-Yu CHEN ; Zhi-Cheng LIN ; Yu-Yu ZHOU ; Tian-Yuan FANG ; Chu-Xia ZHANG ; Biao XIAO ; Kai XIE ; Qing-Juan WANG ; Zhi-Tao LIU ; Li-Ping LI
Progress in Biochemistry and Biophysics 2025;52(2):310-333
		                        		
		                        			
		                        			Alzheimer’s disease (AD) is a central neurodegenerative disease characterized by progressive cognitive decline and memory impairment in clinical. Currently, there are no effective treatments for AD. In recent years, a variety of therapeutic approaches from different perspectives have been explored to treat AD. Although the drug therapies targeted at the clearance of amyloid β-protein (Aβ) had made a breakthrough in clinical trials, there were associated with adverse events. Neuroinflammation plays a crucial role in the onset and progression of AD. Continuous neuroinflammatory was considered to be the third major pathological feature of AD, which could promote the formation of extracellular amyloid plaques and intracellular neurofibrillary tangles. At the same time, these toxic substances could accelerate the development of neuroinflammation, form a vicious cycle, and exacerbate disease progression. Reducing neuroinflammation could break the feedback loop pattern between neuroinflammation, Aβ plaque deposition and Tau tangles, which might be an effective therapeutic strategy for treating AD. Traditional Chinese herbs such as Polygonum multiflorum and Curcuma were utilized in the treatment of AD due to their ability to mitigate neuroinflammation. Non-steroidal anti-inflammatory drugs such as ibuprofen and indomethacin had been shown to reduce the level of inflammasomes in the body, and taking these drugs was associated with a low incidence of AD. Biosynthetic nanomaterials loaded with oxytocin were demonstrated to have the capability to anti-inflammatory and penetrate the blood-brain barrier effectively, and they played an anti-inflammatory role via sustained-releasing oxytocin in the brain. Transplantation of mesenchymal stem cells could reduce neuroinflammation and inhibit the activation of microglia. The secretion of mesenchymal stem cells could not only improve neuroinflammation, but also exert a multi-target comprehensive therapeutic effect, making it potentially more suitable for the treatment of AD. Enhancing the level of TREM2 in microglial cells using gene editing technologies, or application of TREM2 antibodies such as Ab-T1, hT2AB could improve microglial cell function and reduce the level of neuroinflammation, which might be a potential treatment for AD. Probiotic therapy, fecal flora transplantation, antibiotic therapy, and dietary intervention could reshape the composition of the gut microbiota and alleviate neuroinflammation through the gut-brain axis. However, the drugs of sodium oligomannose remain controversial. Both exercise intervention and electromagnetic intervention had the potential to attenuate neuroinflammation, thereby delaying AD process. This article focuses on the role of drug therapy, gene therapy, stem cell therapy, gut microbiota therapy, exercise intervention, and brain stimulation in improving neuroinflammation in recent years, aiming to provide a novel insight for the treatment of AD by intervening neuroinflammation in the future. 
		                        		
		                        		
		                        		
		                        	
8.DIA Proteomics Reveals Mechanism of Acanthopanacis Senticosi Radix et Rhizoma seu Caulis Extract in Treating α-Syn Transgenic Parkinson's Disease in Mice
Qi ZHENG ; Yi LU ; Donghua YU ; Liangyou ZHAO ; Chunsheng LIN ; Fang LU ; Shumin LIU
Chinese Journal of Experimental Traditional Medical Formulae 2025;31(8):40-50
		                        		
		                        			
		                        			ObjectiveTo investigate the mechanism of Acanthopanacis Senticosi Radix et Rhizoma seu Caulis extract (ASH) in treating Parkinson's disease (PD) in mice by Data-Independent Acquisition (DIA) proteomics. MethodsThe α-Synuclein (α-Syn) transgenic PD mice were selected as suitable models for PD, and they were randomly assigned into PD, ASH (61.25 mg·kg-1), and Madopar (97.5 mg·kg-1) groups. Male C57BL/6 mice of the same age were selected as the control group, with eight mice in each group. Mice were administrated with corresponding drugs by gavage once a day for 20 days. The pole climbing time and the number of autonomic activities were recorded to evaluate the exercise ability of mice. Hematoxylin-eosin staining was employed to observe neuronal changes in the substantia nigra of PD mice. Immunohistochemistry (IHC) was employed to measure the tyrosine hydroxylase (TH) activity in the substantia nigra and assess the areal density of α-Syn in the striatum. DIA proteomics was used to compare protein expression in the substantia nigra between groups. IHC was utilized to validate key differentially expressed proteins, including Lactotransferrin, Notch2, Ndrg2, and TMEM 166. The cell counting kit-8 (CCK-8) method was used to investigate the effect of ASH on the viability of PD cells with overexpression of α-Syn. Real-time fluorescence quantitative polymerase chain reaction (Real-time PCR) and Western blot were employed to determine the protein and mRNA levels of Lactotransferrin, Notch2, Ndrg2, and TMEM 166 in PD cells. ResultsCompared with the control group, the model group showed prolonged pole climbing time, diminished coordination ability, reduced autonomic activities (P<0.01), and reduced swelling neurons. Compared with the model group, ASH and Madopar reduced the climbing time, increased autonomic activities (P<0.01), and ameliorated neuronal damage. Compared with the control group, the model group showed a decrease in TH activity in the substantia nigra and an increase in α-Syn accumulation in the striatum (P<0.01). Compared with the model group, the ASH group showed an increase in TH activity and a reduction in α-Syn accumulation (P<0.05). DIA proteomics revealed a total of 464 differentially expressed proteins in the model group compared with the control group, with 323 proteins being up-regulated and 141 down-regulated. A total of 262 differentially expressed proteins were screened in the ASH group compared with the model group, including 85 proteins being up-regulated and 177 down-regulated. Kyoto encylopedia of genes and genomes (KEGG) pathway analysis indicated that ASH primarily regulated the Notch signaling pathway. The model group showed up-regulation in protein levels of Notch2, Ndrg2, and TMEM 166 and down-regulation in the protein level of Lactotransferrin compared with the control group (P<0.01). Compared with the model group, ASH down-regulated the protein levels of Notch2, Ndrg2, and TMEM 166 (P<0.05) while up-regulating the protein level of Lactotransferrin (P<0.01). The IHC results corroborated the proteomics findings. The cell experiment results showed that compared with the control group, the modeling up-regulated the mRNA and protein levels of Notch2, Ndrg2, and TMEM 166 (P<0.01), while down-regulating the mRNA and protein levels of Lactotransferrin (P<0.01). Compared with the model group, ASH reduced the mRNA and protein levels of Notch2, Ndrg2, and TMEM 166 (P<0.01), while increasing the mRNA and protein levels of Lactotransferrin (P<0.05, P<0.01). ConclusionASH may Synergistically inhibit the Notch signaling pathway and mitigate neuronal damage by down-regulating the expression of Notch2 and Ndrg2. Additionally, by up-regulating the expression of Lactotransferrin and down-regulating the expression of TMEM166, ASH can address brain iron accumulation, intervene in ferroptosis, inhibit mitophagy, and mitigate reactive oxygen species damage, thereby protecting nerve cells and contributing to the treatment of PD. 
		                        		
		                        		
		                        		
		                        	
9.Terms Related to The Study of Biomacromolecular Condensates
Ke RUAN ; Xiao-Feng FANG ; Dan LI ; Pi-Long LI ; Yi LIN ; Zheng WANG ; Yun-Yu SHI ; Ming-Jie ZHANG ; Hong ZHANG ; Cong LIU
Progress in Biochemistry and Biophysics 2025;52(4):1027-1035
		                        		
		                        			
		                        			Biomolecular condensates are formed through phase separation of biomacromolecules such as proteins and RNAs. These condensates exhibit liquid-like properties that can futher transition into more stable material states. They form complex internal structures via multivalent weak interactions, enabling precise spatiotemporal regulations. However, the use of inconsistent and non-standardized terminology has become increasingly problematic, hindering academic exchange and the dissemination of scientific knowledge. Therefore, it is necessary to discuss the terminology related to biomolecular condensates in order to clarify concepts, promote interdisciplinary cooperation, enhance research efficiency, and support the healthy development of this field. 
		                        		
		                        		
		                        		
		                        	
10.Mechanism of Huangqi Chifengtang in Treating Atherosclerosis Based on 16S rRNA Sequencing and Metabolomics
Yuqin LIANG ; Jiaqi FU ; Yunhe SHI ; Fang LU ; Donghua YU ; Shumin LIU
Chinese Journal of Experimental Traditional Medical Formulae 2025;31(13):94-103
		                        		
		                        			
		                        			ObjectiveTo investigate the mechanism of action of Huangqi Chifengtang (HQCFT) on rats with atherosclerosis (AS) by regulating the gut microbiota and their metabolites. MethodsA rat model of AS was induced through high-fat diet feeding and vitamin D3 injection, and the modeling lasted for 12 weeks. Fifty eight-week-old male SD rats were randomly divided into five groups: A blank group, a model group, a group receiving a low dose of HQCFT at 1.53 g·kg-1 (HQCFT-L group), a group receiving a high dose of HQCFT at 3.06 g·kg-1 (HQCFT-H group), and a group receiving atorvastatin calcium tablets at 1.8 mg·kg-1 (Ato group), with 10 rats in each group. Oral gavage administration started on the day after model establishment, once daily for four weeks. The efficacy of HQCFT was verified using aortic hematoxylin-eosin (HE) staining and determination of lipid levels and hemorrheology. The real-time polymerase chain reaction (Real-time PCR) was used for detecting inflammatory factor levels in the aorta, high-throughput sequencing for analyzing the gut microbiota composition in intestinal contents, targeted metabolomics for detecting short-chain fatty acid (SCFA) levels, and non-targeted metabolomics for identifying metabolomic profiles of intestinal contents. ResultsCompared with that in the blank group, the aortic tissue of rats in the model group showed significant AS lesions, including endothelial damage, inflammatory infiltration, and formation of fibrous plaques and calcified foci. Moreover, serum triacylglycerol (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C) levels were significantly elevated (P<0.05), while high-density lipoprotein cholesterol (HDL-C) levels were significantly reduced (P<0.05). Significant increases were observed in whole blood viscosity, plasma viscosity, and the mRNA expression levels of NOD-like receptor pyrin domain containing 3 (NLRP3), Caspase-1, interleukin (IL)-β, IL-6, and tumor necrosis factor-α (TNF-α) in aortic tissue (P<0.05). Additionally, gut microbiota composition, SCFA levels, and metabolomic profiles were significantly altered. Compared with those in the model group, serum TC, TG, and LDL-C levels, as well as the whole blood viscosity and plasma viscosity, were significantly reduced in all groups treated with HQCFT (P<0.05). Significant decreases were observed in NLRP3 mRNA expression levels in all groups treated with HQCFT, Caspase-1, IL-β, and IL-6 mRNA expression levels in the HQCFT-H group, and TNF-α mRNA expression levels in the HQCFT-L group (P<0.05). HQCFT reversed the increase in the F/B ratio and dialled back the decrease in the relative abundance of Blautia and the increase in that of Desulfovibrio. HQCFT promoted the production of acetic acid, valeric acid, and propionic acid. Non-targeted metabolomics identified 39 differential metabolites, which were mainly enriched in metabolic pathways such as arachidonic acid metabolism and primary bile acid biosynthesis. ConclusionThe mechanism by which HQCFT ameliorates AS injury may be related to the improvement of dyslipidemia and body inflammatory responses by altering gut microbiota composition, promoting SCFA production, and regulating the levels of metabolites in intestinal contents. 
		                        		
		                        		
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail