1
|
He Y, Shaoyong W, Chen Y, Li M, Gan Y, Sun L, Liu Y, Wang Y, Jin M. The functions of gut microbiota-mediated bile acid metabolism in intestinal immunity. J Adv Res 2025:S2090-1232(25)00307-8. [PMID: 40354934 DOI: 10.1016/j.jare.2025.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/19/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Bile acids, derived from cholesterol in the liver, consist a steroidal core. Primary bile acids and secondary bile acids metabolized by the gut microbiota make up the bile acid pool, which modulate nuclear hormone receptors to regulate immunity. Disruptions in the crosstalk between bile acids and the gut flora are intimately associated with the development and course of gastrointestinal inflammation. AIM OF REVIEW This review provides an extensive summary of bile acid production, transport and metabolism. It also delves into the impact of bile acid metabolism on the body and explores the involvement of bile acid-microbiota interactions in various disease states. Furthermore, the potential of targeting bile acid signaling as a means to prevent and treat inflammatory bowel disease is proposed. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, we primarily address the functions of bile acid-microbiota crosstalk in diseases. Firstly, we summarize bile acid signalling and the factors influencing bile acid metabolism, with highlighting the immune function of microbially conjugated bile acids and the unique roles of different receptors. Subsequently, we emphasize the vital role of bile acids in maintaining a healthy gut microbiota and regulating the intestinal barrier function, energy metabolism and immunity. Finally, we explore differences of bile acid metabolism in different disease states, offering new perspectives on restoring the host's health and the gastrointestinal ecosystem by targeting the gut microbiota-bile acid-bile acid receptor axis.
Collapse
Affiliation(s)
- Yanmin He
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Weike Shaoyong
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Yanli Chen
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Menglin Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yujie Gan
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Lu Sun
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Yalin Liu
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Yizhen Wang
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Mingliang Jin
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China.
| |
Collapse
|
2
|
Ghaffari MH, Ostendorf CS, Hemmert KJ, Schuchardt S, Koch C, Sauerwein H. Longitudinal characterization of plasma and fecal bile acids in dairy heifers from birth to first calving in response to transition milk feeding. J Dairy Sci 2025; 108:5475-5488. [PMID: 40216228 DOI: 10.3168/jds.2025-26307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/27/2025] [Indexed: 05/03/2025]
Abstract
This study aimed to characterize plasma bile acid changes from birth to first calving and evaluate the effects of early transition milk (TM) feeding versus milk replacer (MR) during key stages. Fecal bile acids in TM-fed calves were also analyzed, offering insights into bile acid metabolism. Thirty female Holstein calves were fed TM or MR for the first 5 d, followed by 12 L/d MR. From d 14, calves were fed MR and starter with gradual weaning between wk 8 and 14. Blood samples were collected at 7 time points: 30 min and 12 h after birth, preweaning (wk 2, 6), weaning (wk 14), 8 mo, 13 mo, 3 wk before calving, at calving, and 3 wk after calving. Fecal samples were collected from a subset of TM-fed calves (n = 10) at birth, wk 6, wk 14, 8 mo, and calving. Samples were analyzed for bile acids using the Biocrates MxP Quant 500 kit. Cholic acid (CA) in plasma showed significant time-treatment interactions, with higher levels in TM-fed calves at weaning. Taurine- and glycine-conjugated bile acids had no treatment or time-treatment interactions, but all plasma bile acids showed significant time effects. Principal component analysis revealed that bile acid profiles at birth and after colostrum intake were tightly clustered. Plasma bile acid profiles showed greater dispersion during milk feeding and weaning, with tighter clustering observed postweaning, particularly at 13 mo, and in the transition period. Significant effects were observed for CA, deoxycholic acid (DCA) and chenodeoxycholic acid (CDCA), with CA showing a notable interaction and being higher in TM-fed calves at weaning than in MR-fed calves. Bile acid levels increased toward weaning, peaked at wk 14, and decreased after weaning. Glycine-conjugated bile acids changed over time, with glycocholic acid (GCA) and glycodeoxycholic acid (GDCA) peaking at weaning, and glycochenodeoxycholic acid (GCDCA) being elevated before weaning, decreasing thereafter, and increasing again at calving. Taurine-conjugated bile acids also showed temporal changes, peaking at wk 6. The shifts in bile acid composition from birth to postcalving, with taurolithocholic acid (TLCA), GDCA, and taurocholic acid (TCA) initially dominating, CA increasing at weaning, and GDCA and DCA dominating at calving, with CA increasing again postcalving. During the transition to calving, CA decreased whereas glycine-conjugated bile acids increased relative to taurine-conjugated bile acids in plasma, irrespective of treatment. Fecal bile acid profiles in TM-fed calves clustered distinctly at birth, evolving through pre- to postweaning and calving, with increasing profile overlap over time. Most fecal bile acids, except DCA and CA, were abundant at birth but declined over time. Both DCA and CA increased postweaning, mirroring plasma trends. From wk 6 to calving, DCA was the dominant bile acid, accounting for the highest percentage of total bile acids excreted in feces. Spearman's correlation analysis was performed to assess the relationship between plasma and fecal bile acids in TN-fed calves. A significant positive correlation was observed only for GCDCA (Spearman's rank correlation coefficient [rho] = 0.35), whereas all other bile acids were not correlated. These results illustrate the complex dynamics of bile acid profiles during calf development.
Collapse
Affiliation(s)
- M H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| | - C S Ostendorf
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | - K J Hemmert
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - S Schuchardt
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | - C Koch
- Educational and Research Centre for Animal Husbandry, Hofgut Neumühle, 67728 Münchweiler an der Alsenz, Germany
| | - H Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| |
Collapse
|
3
|
Tao D, Dong Y, Che D, Wang Z, Zheng Y, Han R, Jiang H. Acanthopanax senticosus polysaccharide alleviates LPS-induced intestinal inflammation in piglets by gut microbiota and hyodeoxycholic acid regulation. Int J Biol Macromol 2025; 307:141467. [PMID: 40010458 DOI: 10.1016/j.ijbiomac.2025.141467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 02/28/2025]
Abstract
The purpose of this study is to investigate the effects and mechanisms of Acanthopanax senticosus polysaccharides (ASPS) on lipopolysaccharide (LPS)-induced intestinal injury and growth performance in piglets. Our results indicated that ASPS improved the growth performance in LPS-challenged piglets, including the increase in average daily gain (ADG), average daily feed intake (ADFI), and the feed to gain ratio (F/G). ASPS alleviated LPS-induced intestinal inflammation in piglets, accompanied by the increase in the villus height to crypt depth ratio (VCR) and the decreased in the expression levels of IL-1β, IL-6, and TNF-α. 16S rRNA sequencing results showed that ASPS improved gut microbiota dysbiosis and increased Lactobacillus_sp._L_YJ abundance. The combined analysis of untargeted metabolomics of intestinal contents and serum showed that ASPS significantly increased the levels of hyodeoxycholic acid (HDCA), DHA ethyl ester, and alanylalanine, and the level of HDCA is the highest among all metabolites, suggesting that ASPS regulated the metabolites of intestinal contents and serum to alleviate LPS-induced intestinal inflammation in piglets, and HDCA might play a significant role during this process. Furthermore, we investigated the effects of HDCA on growth performance and intestinal inflammation in LPS-challenged piglets. The results indicated that HDCA alleviated LPS-induced intestinal inflammation and improved the growth performance in piglets. In conclusion, ASPS could alleviate LPS-induced intestinal inflammation in piglets by gut microbiota and hyodeoxycholic acid regulation. These findings might provide strong evidence for ASPS as a feed additive to improve piglet diarrhea, and reveal the therapeutic potential of hyodeoxycholic acid in preventing intestinal inflammation in piglets.
Collapse
Affiliation(s)
- Dapeng Tao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China 130118; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Changchun, China 130118; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Changchun, China 130118
| | - Yangyunyi Dong
- College of Animal Science and Technology, Jilin Agriculture Science and Technology University, Jilin, China 132109
| | - Dongsheng Che
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China 130118; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Changchun, China 130118; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Changchun, China 130118
| | - Zhongshen Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China 130118; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Changchun, China 130118; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Changchun, China 130118
| | - Yingying Zheng
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China 130118; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Changchun, China 130118; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Changchun, China 130118
| | - Rui Han
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China 130118; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Changchun, China 130118; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Changchun, China 130118.
| | - Hailong Jiang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China 130118; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Changchun, China 130118; Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Changchun, China 130118.
| |
Collapse
|
4
|
Ma H, Li R, Qu B, Liu Y, Li P, Zhao J. The Role of Bile Acid in Immune-Mediated Skin Diseases. Exp Dermatol 2025; 34:e70108. [PMID: 40302108 DOI: 10.1111/exd.70108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 04/07/2025] [Accepted: 04/19/2025] [Indexed: 05/01/2025]
Abstract
Immune-mediated skin disorders arise from dysfunctional immune responses, instigating inflammatory dermatoses and a reduced quality of life. The complex pathogenesis likely involves genetic risks, environmental triggers and aberrant immune activation. An emerging body of evidence suggests that bile acid disturbances may critically promote immune pathology in certain skin conditions. Bile acids synthesised from cholesterol regulate nutrient metabolism and immune cell function via nuclear receptors and G protein-coupled receptors (GPCRs). Altered bile acid profiles and receptor expression have been identified in psoriasis, atopic dermatitis (AD) and autoimmune blistering diseases. Disruptions in bile acid signalling affect the inflammatory and metabolic pathways linked to these disorders. Targeting components of the bile acid axis represents a promising therapeutic strategy. This review elucidates the intricate links between bile acid homeostasis and immune dysfunction in inflammatory skin diseases, synthesising evidence that targeting bile acid pathways may unlock innovative therapeutic avenues. This study compiles clinical and experimental data revealing disrupted bile acid signalling and composition in various immune-mediated dermatoses, highlighting the emerging significance of bile acids in cutaneous immune regulation.
Collapse
Affiliation(s)
- Huike Ma
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| | - Ruonan Li
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| | - Baoquan Qu
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| | - Yuchen Liu
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Beijing Institute of Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Li W, Gong X, Niu X, Zhou Y, Ren L, Man Z, Tu P, Xiong X, Liu W, Song Y. Quantitative comparison of bile acid glucuronides sub-metabolome between intrahepatic cholestasis and healthy pregnant women. Anal Bioanal Chem 2025; 417:2823-2835. [PMID: 38990360 DOI: 10.1007/s00216-024-05430-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/15/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Abstract
Because of the pathological indication and the physiological functions, bile acids (BAs) have occupied the research hotspot in recent decades. Although extensive efforts have been paid onto BAs sub-metabolome characterization, as the subfamily, BA glucuronides (gluA-BAs) profile is seldom concerned. Here, we made efforts to develop a LC-MS/MS program enabling quantitative gluA-BAs sub-metabolome characterization and to explore the differential species in serum between intrahepatic cholestasis of pregnancy (ICP) patients and healthy subjects. To gain as many authentic gluA-BAs as possible, liver microsomes from humans, rats, and mice were deployed to conjugate glucuronyl group to authentic BAs through in vitro incubation. Eighty gluA-BAs were captured and subsequently served as authentic compounds to correlate MS/MS spectral behaviors to structural features using squared energy-resolved MS program. Optimal collision energy (OCE) of [M-H]->[M-H-176.1]- was jointly administrated by [M-H]- mass and glucuronidation site, and identical exciting energies corresponding to 50% survival rate of 1st-generation fragment ion (EE50) were observed merely when the aglycone of a gluA-BA was consistent with the suspected structure. Through integrating high-resolution m/z, OCE, and EE50 information to identify gluA-BAs in a BAs pool, 97 ones were found and identified, and further, quantitative program was built for all annotated gluA-BAs by assigning OCEs to [M-H]->[M-H-176.1]- ion transitions. Quantitative gluA-BAs sub-metabolome of ICP was different from that of the healthy group. More GCDCA-3-G, GDCA-3-G, TCDCA-7-G, TDCA-3-G, and T-β-MCA-3-G were distributed in the ICP group. Above all, this study not only offered a promising analytical tool for in-depth gluA-BAs sub-metabolome characterization, but also clarified gluA-BAs allowing the differentiation of ICP and healthy subjects.
Collapse
Affiliation(s)
- Wei Li
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102401, China
| | - Xingcheng Gong
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102401, China
| | - Xiaoya Niu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102401, China
| | - Yuxuan Zhou
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102401, China
| | - Luyao Ren
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102401, China
| | - Zhuo Man
- SCIEX China, Beijing, 100015, China
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102401, China
| | - Xin Xiong
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China.
| | - Wenjing Liu
- School of Pharmacy, Henan University of Chinese Medicine, Jinshui East Road, Zhengdong New District, Zhengzhou, 450046, China.
| | - Yuelin Song
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102401, China.
| |
Collapse
|
6
|
Shen W, Li Z, Wang L, Liu Q, Zhang R, Yao Y, Zhao Z, Ji L. Tumor-resident Malassezia can promote hepatocellular carcinoma development by downregulating bile acid synthesis and modulating tumor microenvironment. Sci Rep 2025; 15:15020. [PMID: 40301518 PMCID: PMC12041395 DOI: 10.1038/s41598-025-99973-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/24/2025] [Indexed: 05/01/2025] Open
Abstract
Bacterial dysbiosis coincides with the carcinogenesis in malignancies such as lung and colon cancer, and has recently been suggested to involve in the pathogenesis of hepatocellular carcinoma (HCC). However, the mycobiome has not yet been definitively linked to liver tumorigenesis. Here we showed that the microbiota composition of HCC tumors was distinct from that of the normal adjacent to tumor (NAT) on the basis of richness and beta-diversity indices. Specifically, the fungal community that infiltrated HCC tumors was markedly enriched for Malassezia spp. and genus Malassezia in tumors was substantially more abundant than that in NAT. We also discovered that the relative abundance of genus Malassezia was strongly correlated with the tumor microenvironment (TME) signatures, including stromal and immune components. In addition, tumor-resident Malassezia could inhibit bile acid synthesis by downregulating the expression level of CYP7 A1 and CYP27 A1. To improve clinical usability, we developed a set of Malassezia-related genes, called Malassezia.Sig, which could accurately predict patient survival. Collectively, our work shows that tumor-resident Malasseiza may promote HCC progression by downregulating bile acid synthesis and modulating the TME, although more studies are needed.
Collapse
Affiliation(s)
- Weixi Shen
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Zhihong Li
- Ganzi Tibetan Autonomous Prefecture People's Hospital, Tibet, 850002, China
| | - Lei Wang
- Women's Health Section, Harbin Red Cross Central Hospital, Harbin, 150076, China
| | - Qi Liu
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Renjie Zhang
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yuhua Yao
- School of Mathematics and Statistics, Hainan Normal University, Haikou, 571158, China
| | - Zhicheng Zhao
- The Fourth Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150018, China.
| | - Lei Ji
- Geneis Beijing Co., Ltd, Beijing, 100102, China.
| |
Collapse
|
7
|
Tao Z, Luo Z, Zou Z, Ye W, Hao Y, Li X, Zheng K, Wu J, Xia J, Zhao Y, Wang Y, Zhang X. Novel insights and an updated review of metabolic syndrome in immune-mediated organ transplant rejection. Front Immunol 2025; 16:1580369. [PMID: 40330480 PMCID: PMC12052740 DOI: 10.3389/fimmu.2025.1580369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Metabolic syndrome (MetS) is a group of symptoms that are characterized by abnormal changes in metabolic substances such as glucose, lipids, proteins, and bile acids. MetS is a common complication after organ transplantation and can further affect the survival and physiological function of the graft by reprograming the patient's immune environment. Additionally, MetS can influence the occurrence of post-transplant complications, such as infections. In recent years, research into the epidemiology and mechanisms of MetS has grown significantly. In this review, we summarize the mechanisms of MetS after transplantation and the mechanisms of hyperglycemia, insulin resistance, hyperlipidemia, abnormal bile acids, and abnormal amino acids on the body's immune cells as related to the effect of metabolic disorders on immune rejection after liver, kidney, heart, skin and other organ transplantation. Finally, we provide an overview of current treatment strategies and offer insights into potential future therapies for managing MetS in transplant recipients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yang Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| | - Yongjun Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Li O, Zhou Y, Kim D, Xu H, Bao Z, Yang F. Lactococcus petauri LZys1 modulates gut microbiota, diminishes ileal FXR-FGF15 signaling, and regulates hepatic function. Microbiol Spectr 2025:e0171624. [PMID: 40243350 DOI: 10.1128/spectrum.01716-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 03/10/2025] [Indexed: 04/18/2025] Open
Abstract
Recent studies have indicated that Lactococcus petauri LZys1 (L. petauri LZys1), isolated from healthy human feces, exhibits a promising probiotic profile in vitro. However, its impact on the physiological status of the host in vivo remains uncertain. The objective of our study was to investigate the effects and mechanisms of orally administering L. petauri LZys1 on gut microbiota and liver function in mice. We administered L. petauri LZys1 through daily oral gavage to C57BL/6 male mice. Subsequently, we analyzed changes in gut microbiota composition using 16S rRNA sequencing and quantified alterations in hepatic-intestinal bile acid (BA) profile. Serum biochemical parameters were assessed to evaluate liver function. Our findings revealed that L. petauri LZys1 led to an increase in body weight, liver mass, and serum aminotransferase levels. Oral administration altered the gut microbiota composition, resulting in reduced diversity and abundance of intestinal bacteria. Additionally, the profiles of BAs were suppressed across organs, associated with the downregulation of the ileum's farnesoid X receptor (FXR)/fibroblast growth factor 15 (FGF15) signaling pathway. The decrease in circulating FGF15 mediated the downregulation of hepatic fibroblast growth factor receptor 4 (FGFR4)/FXR, disrupting BA metabolism and fatty acid oxidation. Our findings suggest that L. petauri LZys1 may impact liver function by influencing the gut microbiota-mediated ileal FXR-FGF15 axis and inhibiting hepatic bile acid metabolism. IMPORTANCE This work elucidated the impact of L. petauri LZys1 on host gut microbiota metabolism and hepatic physiological metabolism. We observed that L. petauri LZys1 administration induced liver weight gain and biochemical parameters changes, in addition to a altered gut microbiota and suppressed bile acid (BA) profiles. Furthermore, we propose that changes in liver status are related to the enterohepatic farnesoid X receptor-fibroblast growth factor axis, which alters bile acid metabolism and disrupts liver function. The above findings suggest that attention should be paid to the effect of probiotics on liver function.
Collapse
Affiliation(s)
- Ouyang Li
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
- Digestive Endoscopy Center, Huadong Hospital, Fudan University, Shanghai, China
| | - Yingshun Zhou
- Department of Pathogenic Biology, Public Center of Experimental Technology of Pathogen Biology Technology Platform, Southwest Medical University, Luzhou, Sichuan, China
| | - Dayoung Kim
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
- Department of Gerontology, Huadong Hospital, Fudan University, Shanghai, China
| | - Han Xu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
- Department of Gerontology, Huadong Hospital, Fudan University, Shanghai, China
| | - Zhijun Bao
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
- Department of Gerontology, Huadong Hospital, Fudan University, Shanghai, China
| | - Fan Yang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
- Department of Gerontology, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Koh YC, Liu CP, Leung SY, Lin WS, Ho PY, Ho CT, Pan MH. Nobiletin Enhances Skeletal Muscle Mass and Modulates Bile Acid Composition in Diet-Induced Obese Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9076-9087. [PMID: 40193085 PMCID: PMC12007094 DOI: 10.1021/acs.jafc.5c00255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/17/2025]
Abstract
Obesity and its associated metabolic disorders─including muscle atrophy─pose significant health challenges, particularly with the increasing prevalence of high-fat diets. This study investigates the effects of nobiletin, a citrus flavonoid, on high-fat-diet-induced obesity-related muscle atrophy and its regulatory role in bile acid metabolism, aiming to determine whether nobiletin supplementation can enhance muscle mass and improve metabolic health in a mouse model. Our findings revealed that nobiletin significantly upregulated CYP7A1 expression in the liver, promoting bile acid synthesis and modulating bile acid composition in the ileum and feces, potentially through microbiota-mediated mechanisms. Furthermore, nobiletin supplementation suppressed muscle atrophy-related proteins, including p-4EBP1, TRIM63, and FBXO32, while promoting the phosphorylation of mTOR/AKT/p70S6K and FOXO3a in skeletal muscle. The FGF15/FGFR4/ERK signaling pathway was notably activated in the skeletal muscle tissues of nobiletin-supplemented mice, suggesting a protective effect against muscle atrophy despite the pathway's inhibition in the liver to promote bile acid synthesis. These results indicate that nobiletin not only mitigates muscle atrophy in the context of obesity but also enhances glucose homeostasis, likely through improved skeletal muscle function. Overall, our study highlights the potential of nobiletin as a therapeutic agent for preventing obesity-related complications, regulating bile acid metabolism, and promoting skeletal muscle health.
Collapse
Affiliation(s)
- Yen-Chun Koh
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Chien-Ping Liu
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Siu-Yi Leung
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Wei-Sheng Lin
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
- Department
of Food Science, National Quemoy University, Quemoy 89250, Taiwan
| | - Pin-Yu Ho
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Chi-Tang Ho
- Department
of Food Science, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Min-Hsiung Pan
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
- Department
of Medical Research, China Medical University
Hospital, China Medical University, Taichung City 40402, Taiwan
| |
Collapse
|
10
|
Jirakran K, Vasupanrajit A, Tunvirachaisakul C, Almulla AF, Kubera M, Maes M. Lipid profiles in major depression, both with and without metabolic syndrome: associations with suicidal behaviors and neuroticism. BMC Psychiatry 2025; 25:379. [PMID: 40234788 PMCID: PMC11998271 DOI: 10.1186/s12888-025-06734-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 03/18/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Significant associations exist between major depressive disorder (MDD), metabolic syndrome (MetS), and cardiovascular disease, potentially attributable to heightened atherogenicity. This study aimed to ascertain if MDD, depression severity, suicidal behaviors, and neuroticism associate with elevated pro-atherogenic indices and reduced anti-atherogenic indices, including a reverse cholesterol transport (RCT) index. METHODS This study comprised 34 healthy controls and 33 MDD patients without MetS, and 35 controls and 31 MDD patients with MetS. It assessed total cholesterol (TC) and free cholesterol (FC), high-density lipoprotein cholesterol (HDLc), low-density lipoprotein cholesterol (LDLc), triglycerides (TG), apolipoprotein (ApoA), ApoB, cholesterol esterification rate, and a RCT composite. RESULTS No significant associations between MDD and lipids were seen in the total study group that combined individuals with and without MetS. In individuals devoid of MetS, MDD is significantly correlated with (a) elevated FC, TG, ApoB, Castelli risk index 1, and ApoB/ApoA, and (b) diminished HDLc, ApoA, and RCT index. In individuals without MetS, there are notable correlations between the severity of depression, suicidal tendencies, neuroticism, and ApoB/ApoA, Castelli risk, and RCT indices. CONCLUSIONS The link between lipids and MDD features cannot be adequately estimated by combining participants with and without MetS. It should be examined in a study sample that excludes subjects with MetS. The depression phenome, suicidal behaviors, and neuroticism correlate with diminished RCT and heightened atherogenicity, which are likely implicated in the pathophysiology of MDD. Increased atherogenicity and lowered RCT may represent novel drug targets for the treatment and prevention of MDD, neuroticism, and suicidal behaviors.
Collapse
Affiliation(s)
- Ketsupar Jirakran
- School of Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the, Thai Red Cross Societyaq , Bangkok, Thailand
- Department of Pediatric, Faculty of Medicine, Center of Excellence for Maximizing Children's Developmental Potential, Chulalongkorn University, Bangkok, Thailand
| | - Asara Vasupanrajit
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the, Thai Red Cross Societyaq , Bangkok, Thailand
| | - Chavit Tunvirachaisakul
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the, Thai Red Cross Societyaq , Bangkok, Thailand.
- Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | - Abbas F Almulla
- School of Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the, Thai Red Cross Societyaq , Bangkok, Thailand
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Marta Kubera
- Department of Experimental Neuroendocrinology, Laboratory of Immunoendocrinology, Polish Academy of Sciences, Maj Institute of Pharmacology, Smetna 12, Krakow, 31-343, Poland
| | - Michael Maes
- School of Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China.
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the, Thai Red Cross Societyaq , Bangkok, Thailand.
- Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
- Research Center, Medical University of Plovdiv, Plovdiv, Bulgaria.
- Research and Innovation Program for the Development of MU - PLOVDIV- (SRIPD-MUP), Creation of a network of research higher schools, National plan for recovery and sustainability, European Union - NextGeneration, EU, Maastricht, Netherlands.
- Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Korea.
| |
Collapse
|
11
|
India Aldana S, Petrick L, Niedzwiecki MM, Valvi D, Just AC, Gutiérrez-Avila I, Kloog I, Barupal DK, Téllez-Rojo MM, Wright RO, Baccarelli AA, Wu H, Colicino E. Pregnancy as a Susceptible Period to Ambient Air Pollution Exposure on the Maternal Postpartum Metabolome. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:6400-6413. [PMID: 40129413 DOI: 10.1021/acs.est.4c10717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Pregnancy is a potential critical window to air pollution exposure for long-term maternal metabolic effects. However, little is known about potential early metabolic mechanisms linking air pollution to maternal metabolic health. We included 544 pregnant Mexican women with both ambient PM2.5 levels during pregnancy and untargeted serum metabolomics to examine associations between pregnancy PM2.5 exposure (overall and monthly) and postpartum metabolites, implementing FDR-adjusted robust linear regression controlling for covariates. Pathway enrichment analyses (in Reactome and MetaboAnalyst) and effect modification by fetal sex and folic acid supplementation were also evaluated. Higher PM2.5 exposure levels throughout pregnancy were associated with higher bile acids and amino acids, dysregulated glycerophospholipids, or lower fatty acyl levels (FDR < 0.05), among other metabolites. Potential critical windows of susceptibility to monthly PM2.5 on metabolites were observed in early to midpregnancy (FDR < 0.005). Main findings were consistent by strata of fetal sex and folic acid supplementation. Metabolic pathways corresponding to positive PM2.5-metabolite associations indicated enriched bile acid, dietary lipid, and transmembrane transport metabolism, whereas for negative PM2.5-metabolite associations, we identified altered pathways involving adipogenesis, incretin peptide hormone, GLP-1, PPAR-alpha, and fatty acid receptors (FDR < 0.05). PM2.5 exposures during pregnancy, especially in early gestation, altered maternal postpartum lipids as well as amino acid metabolism.
Collapse
Affiliation(s)
- Sandra India Aldana
- Department of Environmental Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Lauren Petrick
- Department of Environmental Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Megan M Niedzwiecki
- Department of Environmental Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Damaskini Valvi
- Department of Environmental Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Allan C Just
- Institute at Brown for Environment and Society, Brown University, Providence, Rhode Island 02912, United States
| | - Iván Gutiérrez-Avila
- Department of Environmental Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Itai Kloog
- Department of Environmental Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Dinesh K Barupal
- Department of Environmental Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Martha María Téllez-Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Morelos 62100, Mexico
| | - Robert O Wright
- Department of Environmental Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Andrea A Baccarelli
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, United States
| | - Haotian Wu
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Elena Colicino
- Department of Environmental Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
12
|
Yang Y, Hao C, Jiao T, Yang Z, Li H, Zhang Y, Zhang W, Doherty M, Sun C, Yang T, Li J, Wu J, Zhang M, Wang Y, Xie D, Wang T, Wang N, Huang X, Li C, Gonzalez FJ, Wei J, Xie C, Zeng C, Lei G. Osteoarthritis treatment via the GLP-1-mediated gut-joint axis targets intestinal FXR signaling. Science 2025; 388:eadt0548. [PMID: 40179178 DOI: 10.1126/science.adt0548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/27/2025] [Indexed: 04/05/2025]
Abstract
Whether a gut-joint axis exists to regulate osteoarthritis is unknown. In two independent cohorts, we identified altered microbial bile acid metabolism with reduced glycoursodeoxycholic acid (GUDCA) in osteoarthritis. Suppressing farnesoid X receptor (FXR)-the receptor of GUDCA-alleviated osteoarthritis through intestine-secreted glucagon-like peptide 1 (GLP-1) in mice. GLP-1 receptor blockade attenuated these effects, whereas GLP-1 receptor activation mitigated osteoarthritis. Osteoarthritis patients exhibited a lower relative abundance of Clostridium bolteae, which promoted the formation of ursodeoxycholic acid (UDCA), a precursor of GUDCA. Treatment with C. bolteae and Food and Drug Administration-approved UDCA alleviated osteoarthritis through the gut FXR-joint GLP-1 axis in mice. UDCA use was associated with lower risk of osteoarthritis-related joint replacement in humans. These findings suggest that orchestrating the gut microbiota-GUDCA-intestinal FXR-GLP-1-joint pathway offers a potential strategy for osteoarthritis treatment.
Collapse
Affiliation(s)
- Yuanheng Yang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Cong Hao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Tingying Jiao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China
| | - Zidan Yang
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Bioinformatics Center, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Yuqing Zhang
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- The Mongan Institute, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Weiya Zhang
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham, UK
- Pain Centre Versus Arthritis UK, Nottingham, UK
| | - Michael Doherty
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham, UK
- Pain Centre Versus Arthritis UK, Nottingham, UK
| | - Chuying Sun
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tuo Yang
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Health Management Center, Xiangya Hospital, Central South University, Changsha, China
| | - Jiatian Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Wu
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Mengjiao Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yilun Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Dongxing Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Tingjian Wang
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Huang
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Changjun Li
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jie Wei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- Bioinformatics Center, Furong Laboratory, Changsha, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Basu S, Običan SG, Bertaggia E, Staab H, Izquierdo MC, Gyamfi-Bannerman C, Haeusler RA. Unresolved alterations in bile acid composition and dyslipidemia in maternal and cord blood after UDCA treatment for intrahepatic cholestasis of pregnancy. Am J Physiol Gastrointest Liver Physiol 2025; 328:G364-G376. [PMID: 39947696 PMCID: PMC12053871 DOI: 10.1152/ajpgi.00266.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/25/2024] [Accepted: 02/03/2025] [Indexed: 02/19/2025]
Abstract
Intrahepatic cholestasis of pregnancy (ICP) is characterized by elevated plasma bile acid levels. ICP is linked to adverse metabolic outcomes, including a reported increased risk of gestational diabetes. The standard therapeutic approach for managing ICP is treatment with ursodeoxycholic acid (UDCA) and induction of labor before 40 wk of gestation. To investigate bile acid and metabolic parameters after UDCA treatment, we enrolled 12 ICP patients with singleton pregnancies-half with and half without gestational diabetes-and 7 controls. Our study reveals that after UDCA treatment, notwithstanding a reduction in total bile acid and alanine aminotransferase levels, imbalances persist in the cholic acid (CA) to chenodeoxycholic acid (CDCA) ratio in maternal and cord blood plasma. This indicates a continued dysregulation of bile acid metabolism despite therapeutic intervention. Maternal plasma lipid analysis showed a distinct maternal dyslipidemia pattern among patients with ICP, marked by elevated cholesterol levels on VLDL particles and heightened triglyceride concentrations on LDL particles, persisting even after UDCA treatment. Cord plasma lipid profiles in patients with ICP exhibited elevated triglyceride and free fatty acid levels alongside a tendency toward increased β-hydroxybutyrate. The changes in lipid metabolism in both maternal and cord blood correlated with the high CA/CDCA ratio but not total bile acid levels or gestational diabetes status. Understanding the imbalances in maternal and cord bile acid and lipid profiles that persist after standard UDCA therapy provides insights for improving management strategies and mitigating the long-term consequences of ICP.NEW & NOTEWORTHY This study uncovers that despite ursodeoxycholic acid treatment, intrahepatic cholestasis of pregnancy (ICP) is associated with increases in the ratio of cholic acid to chenodeoxycholic acid in both maternal and cord blood, suggesting ongoing dysregulation of bile acid metabolism. The high cholic to chenodeoxycholic acid ratio is correlated with maternal dyslipidemia and high cord blood lipids. These findings may inform more targeted approaches to managing ICP.
Collapse
Affiliation(s)
- Srijani Basu
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, United States
- Columbia University Digestive and Liver Disease Research Center, Columbia University, New York, New York, United States
- Department of Medicine, Columbia University, New York, New York, United States
| | - Sarah G Običan
- Department of Obstetrics and Gynecology, Columbia University, New York, New York, United States
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, Florida, United States
| | - Enrico Bertaggia
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, United States
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States
| | - Hannah Staab
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, United States
| | - M Concepcion Izquierdo
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, United States
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States
| | | | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, United States
- Columbia University Digestive and Liver Disease Research Center, Columbia University, New York, New York, United States
- Department of Medicine, Columbia University, New York, New York, United States
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States
| |
Collapse
|
14
|
Zeng W, Sun M, Cao J, Chen C, Jiang S, Wang Y, Yang W, Zhao Z, Jin J. Triterpenoids from ilicis rotundae cortex ameliorate hyperlipidemia by affecting bile acids-hepatointestinal FXR axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156537. [PMID: 40023069 DOI: 10.1016/j.phymed.2025.156537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/15/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Hyperlipidemia is a lipid metabolism disorder that, in severe cases, can lead to conditions such as hypertension, coronary heart disease, and cirrhosis. Previous studies have identified Ilicis Rotundae Cortex (IRC) crude extract as having the potential to regulate blood lipids. However, whether the triterpenoids therein are the principal agents responsible for hypolipidemic effects and their specific mechanisms of action remain unexplored. This study aimed to investigate the effects of total triterpenoids (TT) extract derived from IRC on hyperlipidemia and to elucidate their potential mechanisms. METHODS TT extract was first prepared and characterized to assess their hypolipidemic activity in cell models. A hyperlipidemia mouse model was established by using C57BL/6 J mice fed a high-fat, high-sugar, and high-cholesterol diet for 8 weeks. TT extract was administered as a prophylactic intervention for 4 weeks to evaluate its impact on blood lipid levels, liver lipid metabolism, and liver function. Based on progressive analysis, this study integrated serum non-targeted metabolomics analysis strategy and bile acids-targeted metabolomics analysis strategy. It was combined with modern molecular biology techniques to reveal the mechanism by which TT extract ameliorated the symptoms of hyperlipidemia through a cascade approach. RESULTS TT extract treatment significantly reduced lipid levels in hyperlipidemic mice. Notably, TT extract down-regulated bile acid levels, particularly bile acids as FXR antagonists such as T-β-MCA, β-MCA, TUDCA, and UDCA. This effect is likely mediated through alterations in the hepatic FXR-SHP and ileal FXR-FGF15 signaling pathways. TT extract administration led to decreased expression of CYP7A1 and CYP7B1, resulting in reduced bile acid levels in vivo. Additionally, FXR expression was upregulated in both the liver and ileum, potentially activating FGF15 in the ileum, which in turn transmits signals to the liver and modulates SHP and BSEP expression. These changes contribute to the regulation of bile acid synthesis, metabolism, and excretion. In vitro experiments also demonstrated that TT extract influenced the protein expression of FXR and FGF19. CONCLUSION Our findings demonstrate that TT extract from IRC has hypolipidemic effects. This study is the first to reveal the mechanism by which TT extract improves hyperlipidemia from the perspective of the hepatic-intestinal axis and bile acid metabolism. Its underlying mechanism is related to activating the intestinal FXR-FGF15/19 signaling pathway, which transmits signals to the liver, thereby affecting the hepatic FXR-SHP signaling pathway. This results in improved bile acid metabolism, ultimately reducing hepatic injury and ileal inflammation to exert hypolipidemic effects.
Collapse
Affiliation(s)
- Wei Zeng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China; Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, 519000, PR China
| | - Mengjia Sun
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China; Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, 519000, PR China
| | - Jiamin Cao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China; Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, 519000, PR China
| | - Caixin Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China; Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, 519000, PR China
| | - Shiqin Jiang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Yuanyuan Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China; Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, 519000, PR China
| | - Weiqun Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China; Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, 519000, PR China
| | - Zhongxiang Zhao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China; Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, 519000, PR China.
| | - Jing Jin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, PR China.
| |
Collapse
|
15
|
Kim WJ, Ryu R, Doo EH, Choi Y, Kim K, Kim BK, Kim H, Kim M, Huh CS. Supplementation with the Probiotic Strains Bifidobacterium longum and Lactiplantibacillus rhamnosus Alleviates Glucose Intolerance by Restoring the IL-22 Response and Pancreatic Beta Cell Dysfunction in Type 2 Diabetic Mice. Probiotics Antimicrob Proteins 2025; 17:541-556. [PMID: 37804432 DOI: 10.1007/s12602-023-10156-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2023] [Indexed: 10/09/2023]
Abstract
Type 2 diabetes (T2D) is known as adult-onset diabetes, but recently, T2D has increased in the number of younger people, becoming a major clinical burden in human society. The objective of this study was to determine the effects of Bifidobacterium and Lactiplantibacillus strains derived from the feces of 20 healthy humans on T2D development and to understand the mechanism underlying any positive effects of probiotics. We found that Bifidobacterium longum NBM7-1 (Chong Kun Dang strain 1; CKD1) and Lactiplantibacillus rhamnosus NBM17-4 (Chong Kun Dang strain 2; CKD2) isolated from the feces of healthy Korean adults (n = 20) have anti-diabetic effects based on the insulin sensitivity. During the oral gavage for 8 weeks, T2D mice were supplemented with anti-diabetic drugs (1.0-10 mg/kg body weight) to four positive and negative control groups or four probiotics (200 uL; 1 × 109 CFU/mL) to groups separately or combined to the four treatment groups (n = 6 per group). While acknowledging the relatively small sample size, this study provides valuable insights into the potential benefits of B. longum NBM7-1 and L. rhamnosus NBM17-4 in mitigating T2D development. The animal gene expression was assessed using a qRT-PCR, and metabolic parameters were assessed using an ELISA assay. We demonstrated that B. longum NBM7-1 in the CKD1 group and L. rhamnosus NBM17-4 in the CKD2 group alleviate T2D development through the upregulation of IL-22, which enhances insulin sensitivity and pancreatic functions while reducing liver steatosis. These findings suggest that B. longum NBM7-1 and L. rhamnosus NBM17-4 could be the candidate probiotics for the therapeutic treatments of T2D patients as well as the prevention of type 2 diabetes.
Collapse
Affiliation(s)
- Won Jun Kim
- Department of Agricultural Biotechnology, College of Agriculture Sciences, Seoul National University, Seoul, South Korea
| | - Ri Ryu
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang, South Korea
| | - Eun-Hee Doo
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang, South Korea
- Department of Yuhan Biotechnology, School of Bio-Health Sciences, Yuhan University, Bucheon, 14780, South Korea
| | - Yukyung Choi
- Research Institute, Chong Kun Dang Bio Co. Ltd, Ansan, South Korea
| | - Kyunghwan Kim
- Research Institute, Chong Kun Dang Bio Co. Ltd, Ansan, South Korea
| | - Byoung Kook Kim
- Research Institute, Chong Kun Dang Bio Co. Ltd, Ansan, South Korea
| | - Heebal Kim
- Department of Agricultural Biotechnology, College of Agriculture Sciences, Seoul National University, Seoul, South Korea
- Department of Animal Science and Biotechnology, Seoul National University, Seoul, South Korea
| | - Myunghoo Kim
- Department of Animal Science, Pusan National University, Miryang, South Korea.
| | - Chul Sung Huh
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang, South Korea.
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, South Korea.
| |
Collapse
|
16
|
Zhou Z, Xu D, Huang L, Cui Y, Chen H, Tang J. Farnesoid X Receptor Regulated Sepsis-Induced Abnormal Bile Acid Metabolism via the Fibroblast Growth Factor 15/Fibroblast Growth Factor Receptor 4 Pathway. Immun Inflamm Dis 2025; 13:e70155. [PMID: 40192065 PMCID: PMC11973727 DOI: 10.1002/iid3.70155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/06/2025] [Accepted: 01/30/2025] [Indexed: 04/10/2025] Open
Abstract
OBJECTIVE The study aims to investigate the mechanism of Farnesoid X receptor (FXR) activation in sepsis-induced abnormal bile acid metabolism and the metabolism status of each bile acid type. METHODS The sepsis mouse model was developed via lipopolysaccharide intraperitoneal injection and confirmed via hematoxylin and eosin (H&E) staining. FXR agonist activated the FXR/fibroblast growth factor (FGF)15/FGFR pathway via quantitative real-time polymerase chain reaction and Western blot. Consequently, metabolomics and bioinformatics analysis were conducted to identify the alterations in each kind of bile acid content following FXR agonist/inhibitor intervention. RESULTS The H&E staining indicated that FXR activation alleviates the liver injury of the sepsis mouse model. The increased FGF15 and FXFR expression levels and decreased CYP7A1 demonstrated FXR/FGF15/FGFR pathway activation following FXR agonist treatment. Furthermore, total bile acid, interleukin (IL)-6, and tumor necrosis factor-α concentrations were downregulated after FXR activation, whereas IL-10 concentration was upregulated, indicating the alleviated effect of FXR agonist in sepsis. Consequently, metabolomics and bioinformatics analysis determined that T-a-MCA were downregulated in both FXR agonist and inhibitor groups, whereas six bile acid types were altered in the control group. CONCLUSION FXR activation was crucial in alleviating sepsis-induced hepatic injury and cholestasis through the FGF15/FGFR signaling pathway, and FXR may act as a potential preventive and intervention target of sepsis.
Collapse
Affiliation(s)
- Ziyang Zhou
- Trauma‐Emergency & Critical Care Medicine CenterShanghai Fifth People's Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Dan Xu
- Trauma‐Emergency & Critical Care Medicine CenterShanghai Fifth People's Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Liou Huang
- Trauma‐Emergency & Critical Care Medicine CenterShanghai Fifth People's Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Yuhui Cui
- Trauma‐Emergency & Critical Care Medicine CenterShanghai Fifth People's Hospital Affiliated to Fudan UniversityShanghaiChina
| | - Hui Chen
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life ScienceEast China Normal UniversityShanghaiChina
| | - Jianguo Tang
- Trauma‐Emergency & Critical Care Medicine CenterShanghai Fifth People's Hospital Affiliated to Fudan UniversityShanghaiChina
| |
Collapse
|
17
|
Liu J, Guo M, Yuan X, Fan X, Wang J, Jiao X. Gut Microbiota and Their Metabolites: The Hidden Driver of Diabetic Nephropathy? Unveiling Gut Microbe's Role in DN. J Diabetes 2025; 17:e70068. [PMID: 40189872 PMCID: PMC11973130 DOI: 10.1111/1753-0407.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/21/2025] [Accepted: 02/17/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a severe microvascular complication of diabetes with a complex pathogenesis. METHODS Recent studies were reviewed to explore the role of gut microbiota and its metabolites in DN development. RESULTS Dysbiosis of gut bacteria contributes to pathological changes such as glomerular sclerosis and renal tubule injury. Microbial metabolites are involved in DN through immune and inflammatory pathways. CONCLUSIONS Understanding the relationship between gut microbiota, its metabolites, and DN may offer potential implications for DN diagnosis, prevention, and treatment. Translating this knowledge into clinical practice presents challenges and opportunities.
Collapse
Affiliation(s)
- Jinzhou Liu
- Department of PhysiologyThe Key Laboratory of Physiology of Shanxi Province, the Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical UniversityTaiyuanChina
| | - Min Guo
- Department of PhysiologyThe Key Laboratory of Physiology of Shanxi Province, the Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical UniversityTaiyuanChina
| | - Xiaobin Yuan
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Xiao Fan
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Jin Wang
- Department of PhysiologyThe Key Laboratory of Physiology of Shanxi Province, the Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical UniversityTaiyuanChina
| | - Xiangying Jiao
- Department of PhysiologyThe Key Laboratory of Physiology of Shanxi Province, the Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
18
|
Schnabl B, Damman CJ, Carr RM. Metabolic dysfunction-associated steatotic liver disease and the gut microbiome: pathogenic insights and therapeutic innovations. J Clin Invest 2025; 135:e186423. [PMID: 40166938 PMCID: PMC11957707 DOI: 10.1172/jci186423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major cause of liver disease worldwide, and our understanding of its pathogenesis continues to evolve. MASLD progresses from steatosis to steatohepatitis, fibrosis, and cirrhosis, and this Review explores how the gut microbiome and their metabolites contribute to MASLD pathogenesis. We explore the complexity and importance of the intestinal barrier function and how disruptions of the intestinal barrier and dysbiosis work in concert to promote the onset and progression of MASLD. The Review focuses on specific bacterial, viral, and fungal communities that impact the trajectory of MASLD and how specific metabolites (including ethanol, bile acids, short chain fatty acids, and other metabolites) contribute to disease pathogenesis. Finally, we underscore how knowledge of the interaction between gut microbes and the intestinal barrier may be leveraged for MASLD microbial-based therapeutics. Here, we include a discussion of the therapeutic potential of prebiotics, probiotics, postbiotics, and microbial-derived metabolites.
Collapse
Affiliation(s)
- Bernd Schnabl
- Department of Medicine, Division of Gastroenterology, UCSD, San Diego, California, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, California, USA
| | - Christopher J. Damman
- Department of Medicine, Division of Gastroenterology, University of Washington, Seattle, Washington, USA
| | - Rotonya M. Carr
- Department of Medicine, Division of Gastroenterology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
19
|
Zhu ZM, Liu HY, An N, Li AL, Li J, Wang SJ, Yang G, Duan YW, Yang Y, Zhang M, Zhu QF, Liu SM, Feng YQ. Metabolic Profiling Reveals Potential Prognostic Biomarkers for SFTS: Insights into Disease Severity and Clinical Outcomes. Metabolites 2025; 15:228. [PMID: 40278357 PMCID: PMC12028903 DOI: 10.3390/metabo15040228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Severe fever with thrombocytopenia syndrome (SFTS) is a viral infection primarily found in Asia, with a case fatality rate of about 10%. Despite its increasing prevalence, the underlying pathogenic mechanisms remain poorly understood, limiting the development of effective therapeutic interventions. Methods: We employed an untargeted metabolomics approach using liquid chromatography-mass spectrometry (LC-MS) to analyze serum samples from 78 SFTS patients during the acute phase of their illness. Differential metabolic features between survival and fatal cases were identified through multivariate statistical analysis. Furthermore, we constructed a metabolic prognostic model based on these biomarkers to predict disease severity. Results: Significant alterations were observed in four key metabolic pathways: sphingolipid metabolism, biosynthesis of phenylalanine, tyrosine, and tryptophan, primary bile acid biosynthesis, and phenylalanine metabolism. Elevated levels of phenyllactic acid and isocitric acid were strongly associated with adverse outcomes and demonstrated high discriminatory power in distinguishing fatal cases from survivors. The metabolic prognostic model incorporating these biomarkers achieved a sensitivity of 75% and a specificity of 90% in predicting disease severity. Conclusions: Our findings highlight the pivotal role of metabolic dysregulation in the pathogenesis of SFTS and suggest that targeting specific metabolic pathways could open new avenues for therapeutic development. The identification of prognostic biomarkers provides a valuable tool for early risk stratification and timely clinical intervention, potentially improving patient outcomes.
Collapse
Affiliation(s)
- Zhuo-Min Zhu
- School of Bioengineering and Health, Wuhan Textile University, Wuhan 430200, China; (Z.-M.Z.); (N.A.); (J.L.); (Y.-Q.F.)
- School of Public Health, Wuhan University, Wuhan 430072, China;
| | - Huan-Yu Liu
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (H.-Y.L.); (A.-L.L.); (G.Y.); (Y.-W.D.); (Y.Y.)
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Na An
- School of Bioengineering and Health, Wuhan Textile University, Wuhan 430200, China; (Z.-M.Z.); (N.A.); (J.L.); (Y.-Q.F.)
- School of Public Health, Wuhan University, Wuhan 430072, China;
| | - An-Ling Li
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (H.-Y.L.); (A.-L.L.); (G.Y.); (Y.-W.D.); (Y.Y.)
| | - Jia Li
- School of Bioengineering and Health, Wuhan Textile University, Wuhan 430200, China; (Z.-M.Z.); (N.A.); (J.L.); (Y.-Q.F.)
- School of Public Health, Wuhan University, Wuhan 430072, China;
| | - Sai-Jun Wang
- School of Public Health, Wuhan University, Wuhan 430072, China;
| | - Gui Yang
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (H.-Y.L.); (A.-L.L.); (G.Y.); (Y.-W.D.); (Y.Y.)
| | - Yong-Wei Duan
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (H.-Y.L.); (A.-L.L.); (G.Y.); (Y.-W.D.); (Y.Y.)
| | - Ying Yang
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (H.-Y.L.); (A.-L.L.); (G.Y.); (Y.-W.D.); (Y.Y.)
| | - Mei Zhang
- Department of Clinical Laboratory, Ezhou Hospital of Traditional Chinese Medicine, Ezhou 436000, China;
| | - Quan-Fei Zhu
- School of Bioengineering and Health, Wuhan Textile University, Wuhan 430200, China; (Z.-M.Z.); (N.A.); (J.L.); (Y.-Q.F.)
| | - Song-Mei Liu
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (H.-Y.L.); (A.-L.L.); (G.Y.); (Y.-W.D.); (Y.Y.)
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yu-Qi Feng
- School of Bioengineering and Health, Wuhan Textile University, Wuhan 430200, China; (Z.-M.Z.); (N.A.); (J.L.); (Y.-Q.F.)
- School of Public Health, Wuhan University, Wuhan 430072, China;
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| |
Collapse
|
20
|
Romeo M, Dallio M, Di Nardo F, Napolitano C, Vaia P, Martinelli G, Federico P, Olivieri S, Iodice P, Federico A. The Role of the Gut-Biliary-Liver Axis in Primary Hepatobiliary Liver Cancers: From Molecular Insights to Clinical Applications. J Pers Med 2025; 15:124. [PMID: 40278303 PMCID: PMC12028696 DOI: 10.3390/jpm15040124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/26/2025] Open
Abstract
Background: Hepatobiliary liver cancers (HBLCs) represent the sixth most common neoplasm in the world. Hepatocellular carcinoma (HCC) and cholangiocarcinoma (CC) constitute the main HBLC types, with alarming epidemiological projections. Methods: In recent decades, alterations in gut microbiota, with mutual implications on the gut-liver axis and gut-biliary axis permeability status, have been massively investigated and proposed as HBLC pathogenetic deus ex machina. Results: In the HCC setting, elevated intestinal levels of Escherichia coli and other Gram-negative bacteria have been demonstrated, resulting in a close association with increased lipopolysaccharide (LPS) serum levels and, consequently, chronic systemic inflammation. In contrast, the intestinal microbiota of HCC individuals feature reduced levels of Lactobacillus spp., Bifidobacterium spp., and Enterococcus spp. In the CC setting, evidence has revealed an increased expression of Lactobacillus spp., with enhanced levels of Actynomices spp. and Alloscardovia spp. Besides impaired strains/species representation, gut-derived metabolites, including bile acids (BAs), short-chain fatty acids (SCFAs), and oxidative-stress-derived products, configure a network severely impacting the progression of HBLC. Conclusions: In the era of Precision Medicine, the clarification of microbiota composition and functioning in HCC and CC settings can contribute to the identification of individual signatures, potentially providing novel diagnostic markers, therapeutic approaches, and prognostic/predictive tools.
Collapse
Affiliation(s)
- Mario Romeo
- Department of Precision Medicine, Hepatogastroenterology Division, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (F.D.N.); (C.N.); (P.V.); (G.M.); (S.O.); (A.F.)
| | - Marcello Dallio
- Department of Precision Medicine, Hepatogastroenterology Division, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (F.D.N.); (C.N.); (P.V.); (G.M.); (S.O.); (A.F.)
| | - Fiammetta Di Nardo
- Department of Precision Medicine, Hepatogastroenterology Division, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (F.D.N.); (C.N.); (P.V.); (G.M.); (S.O.); (A.F.)
| | - Carmine Napolitano
- Department of Precision Medicine, Hepatogastroenterology Division, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (F.D.N.); (C.N.); (P.V.); (G.M.); (S.O.); (A.F.)
| | - Paolo Vaia
- Department of Precision Medicine, Hepatogastroenterology Division, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (F.D.N.); (C.N.); (P.V.); (G.M.); (S.O.); (A.F.)
| | - Giuseppina Martinelli
- Department of Precision Medicine, Hepatogastroenterology Division, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (F.D.N.); (C.N.); (P.V.); (G.M.); (S.O.); (A.F.)
| | - Pierluigi Federico
- Pharmaceutical Department, ASL NA3 Sud, Torre del Greco, 80059 Naples, Italy;
| | - Simone Olivieri
- Department of Precision Medicine, Hepatogastroenterology Division, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (F.D.N.); (C.N.); (P.V.); (G.M.); (S.O.); (A.F.)
| | | | - Alessandro Federico
- Department of Precision Medicine, Hepatogastroenterology Division, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (F.D.N.); (C.N.); (P.V.); (G.M.); (S.O.); (A.F.)
| |
Collapse
|
21
|
Luo T, Zhao X, Jiang X, Li P, Gu X, Xie X, Liu H. Serum metabolomic analysis in patients with Hashimoto's thyroiditis positive for TgAb or TPOAb: a preliminary study. Sci Rep 2025; 15:9945. [PMID: 40121266 PMCID: PMC11929742 DOI: 10.1038/s41598-025-90467-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Hashimoto's thyroiditis (HT) is a prevalent autoimmune disorder, yet the metabolic abnormalities associated with HT and their relationship to antibody positivity remain poorly understood. This study aimed to characterize the distinct metabolic profiles associated with thyroid peroxidase antibody (TPOAb) and thyroglobulin antibody (TgAb) positivity in female patients with HT. Serum metabolomic analysis was performed on 14 TPOAb-positive patients, 4 TgAb-positive patients, and 14 sex-matched healthy controls, evaluating a total of 225 metabolites. Partial least squares discriminant analysis (PLS-DA) revealed significant metabolic differences among the groups, identifying 36 key metabolites. Of these, 13 metabolites showed significant differences between the TPOAb-positive group and healthy controls, while 23 metabolites exhibited marked differences between the TgAb-positive group and controls. Further correlation analysis revealed a moderate positive association between TgAb and phenylacetyl-L-glutamine, while TPOAb was strongly correlated with LPC 16:0 sn-1. Additionally, metabolic pathway analysis showed significant activation of glycine, serine, and threonine metabolism in the TPOAb-positive group, whereas the TgAb-positive group exhibited enhanced activity in galactose metabolism. These findings suggest that TPOAb and TgAb positivity are associated with distinct metabolic profiles, reflecting their differential roles in metabolic pathways linked to Hashimoto's thyroiditis. This study provides valuable exploratory evidence of metabolic abnormalities in HT under different antibody-positive states, laying the foundation for future large-scale investigations to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Tao Luo
- Endocrinology and Metabolism Department, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Xinyu Zhao
- Endocrinology and Metabolism Department, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Xiao Jiang
- Endocrinology and Metabolism Department, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Pengqian Li
- Endocrinology and Metabolism Department, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Xiaotong Gu
- Endocrinology and Metabolism Department, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Xingjie Xie
- Endocrinology and Metabolism Department, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Haixia Liu
- Endocrinology and Metabolism Department, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China.
| |
Collapse
|
22
|
Tang J, Xu W, Yu Y, Yin S, Ye BC, Zhou Y. The role of the gut microbial metabolism of sterols and bile acids in human health. Biochimie 2025; 230:43-54. [PMID: 39542125 DOI: 10.1016/j.biochi.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Sterols and bile acids are vital signaling molecules that play key roles in systemic functions, influencing the composition of the human gut microbiota, which maintains a symbiotic relationship with the host. Additionally, gut microbiota-encoded enzymes catalyze the conversion of sterols and bile acids into various metabolites, significantly enhancing their diversity and biological activities. In this review, we focus on the microbial transformations of sterols and bile acids in the gut, summarize the relevant bacteria, genes, and enzymes, and review the relationship between the sterols and bile acids metabolism of gut microbiota and human health. This review contributes to a deeper understanding of the crucial roles of sterols and bile acids metabolism by gut microbiota in human health, offering insights for further investigation into the interactions between gut microbiota and the host.
Collapse
Affiliation(s)
- Jiahui Tang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Wenwu Xu
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yangfan Yu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Shengxiang Yin
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Bang-Ce Ye
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Yunyan Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.
| |
Collapse
|
23
|
Wang J, Zhong MY, Liu YX, Yu JY, Wang YB, Zhang XJ, Sun HP. Branched-chain amino acids promote hepatic Cyp7a1 expression and bile acid synthesis via suppressing FGF21-ERK pathway. Acta Pharmacol Sin 2025; 46:662-671. [PMID: 39567750 PMCID: PMC11845675 DOI: 10.1038/s41401-024-01417-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024]
Abstract
Branched-chain amino acids (BCAAs) including leucine, isoleucine and valine have been linked with metabolic and cardiovascular diseases. BCAAs homeostasis is tightly controlled by their catabolic pathway. BCKA dehydrogenase (BCKD) complex is the rate-limiting step for BCAA catabolism. Mitochondrial phosphatase 2C (PP2Cm) dephosphorylates the BCKD E1alpha subunit and activates BCKD complex. Deficiency of PP2Cm impairs BCAA catabolism, leading to higher plasma BCAA concentrations. Emerging evidence shows that bile acids are key regulators of glucose, lipid and energy metabolism. In this study, we investigated whether a direct link existed between BCAAs and bile acids metabolism. Wild-type mice were fed with normal-BCAA or high-BCAA diet, while PP2Cm deficiency mice were fed with normal chow for 14 weeks. The mice were fasted for 6 h before tissue harvest to exclude metabolic changes due to immediate food intake. We showed that the bile acids in tissues and feces were significantly elevated in wild-type mice fed with high-BCAA diet as well as in PP2Cm deficiency mice fed with normal chow. These mice displayed significantly increased expression of cholesterol 7 alpha-hydroxylase (CYP7A1), the rate-limiting enzyme of bile acid synthesis in liver, and 7α-hydroxy-4-cholesten-3-one (C4), a freely diffusible metabolite downstream of CYP7A1 in plasma. BCAAs induced Cyp7a1 expression in cultured hepatocytes. In mouse liver and cultured hepatocytes, we demonstrated that elevated BCAAs inhibited fibroblast growth factor 21 (FGF21) expression and ERK signaling pathway. Direct inhibition of ERK by U0126 (800 nM) markedly induced Cyp7a1 expression in cultured hepatocytes. Moreover, the induced Cyp7a1 expression and inhibitory effects of BCAAs on ERK signaling pathway were abolished by treatment with recombinant FGF21 protein in mouse liver and cultured hepatocytes. Collectively, this study demonstrates a direct link between BCAAs and bile acid synthesis. BCAAs promotes Cyp7a1 expression and bile acid synthesis in liver via inhibiting FGF21-ERK signaling pathway. BCAAs-regulated bile acid synthesis and homeostasis may contribute to developing novel therapeutic strategies for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Ji Wang
- Department of Clinical Laboratory, The Second People's Hospital of Hefei / Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, China
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Meng-Yu Zhong
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Yun-Xia Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- The Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Jia-Yu Yu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yi-Bin Wang
- The Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Xue-Jiao Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| | - Hai-Peng Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
24
|
Fang Q, Hou X, Fan L, Deng Y, Li X, Zhang H, Wang H, Fu Z, Sun B, Shu X, Du H, Liu Y. Gut microbiota derived DCA enhances FOLFOX efficacy via Ugt1a6b mediated enterohepatic circulation in colon cancer. Pharmacol Res 2025; 213:107636. [PMID: 39894186 DOI: 10.1016/j.phrs.2025.107636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
FOLFOX (5-Fluorouracil, Calcium Folinate combined with Oxaliplatin) is a preferred chemotherapy regimen for colon cancer, but its limited efficacy remains a major challenge, significantly impairs patient outcomes. There is an urgent need to identify strategies to improve its therapeutic effectiveness. Our previous studies have suggested that gut microbiota-derived bile acids may be involved in the anticancer effect of FOLFOX in vitro, however, the underlying mechanism remains unclear. In this study, we investigated the role of bile acids in modulating FOLFOX efficacy and the related mechanisms. We first demonstrated that bile acids depletion (cholestyramine treatment) enhanced FOLFOX efficacy in an orthotopic colon cancer mouse model, suggesting that bile acids play a key role in FOLFOX's therapeutic effects. Further, based on the system screen of 15 bile acids on FOLFOX efficacy via MTT, colony formation and flow cytometry assay, Deoxycholic Acid (DCA) and Glycodeoxycholic Acid (GDCA) were annotated as potential modulators of FOLFOX efficacy. Among these, DCA was further validated to significantly enhance FOLFOX's anti-colon cancer effects in vivo. Transcriptomic analysis and subsequent biological experiments revealed that DCA enhanced FOLFOX efficacy via Ugt1a6b. In conclusion, our findings establish that gut microbiota-derived DCA enhances the efficacy of FOLFOX potentially via Ugt1a6b mediated enterohepatic circulation, providing novel insights into a synergistic therapeutic strategy for improving colon cancer treatment.
Collapse
Affiliation(s)
- Qian Fang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Xiaoying Hou
- Hubei Key Laboratory of Cognitive and Affective Disorders, Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Limei Fan
- Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Yufei Deng
- Hubei Key Laboratory of Cognitive and Affective Disorders, Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Xiaoxuan Li
- Hubei Key Laboratory of Cognitive and Affective Disorders, Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Hongyun Zhang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Haiping Wang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Zhengqi Fu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Binlian Sun
- Hubei Key Laboratory of Cognitive and Affective Disorders, Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Xiji Shu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Hongzhi Du
- Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China; Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan 430065, China.
| | - Yuchen Liu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
25
|
Xu Y, Chen Y, Zhang J, Wang J, Yang S, Zhao H, Wu L, Lei J, Zhou Y, Peng J, Jiang L, Chen Q, Xin R, Zhou J, Li Y. RNASET2 Deficiency Induces Hepatocellular Carcinoma Metastasis through Cholesterol-Triggered MET Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411888. [PMID: 39903758 PMCID: PMC11948071 DOI: 10.1002/advs.202411888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/21/2024] [Indexed: 02/06/2025]
Abstract
Metastasis remains a significant challenge in the treatment of hepatocellular carcinoma (HCC). The role of ribonuclease T2 (RNASET2) in HCC is still uncertain, although it has been reported to have contradictory effects on some cancers. Here, it is demonstrated that RNASET2 knockout leads to significant accumulation of cholesterol, which in turn promotes MET-mediated HCC metastasis. Mechanistically, the absence of RNASET2 hinders the degradation of RNA into uridine, thereby reducing the conversion to UTP. This reduction restrains glucuronate metabolism and the expression of the related enzyme UDP-glucuronosyltransferase (UGT)1A1, ultimately resulting in the accumulation of cholesterol due to decreased formation of glucuronidated-bile acids. The administration of cholesterol induces the migration and invasion of HCC cells through MET (mesenchymal-epithelial transition factor) activation. However, the deficiency of RNASET2-induced HCC metastasis can be reversed by blocking MET with shRNA or savolitinib. The study identifies RNASET2 as a key regulator that coordinates RNA, glucuronate, and cholesterol metabolism. Its deficiency drives HCC metastasis through cholesterol-triggered MET activation. These findings highlight the potential of targeting RNASET2 and MET in improving the prognosis of HCC.
Collapse
Affiliation(s)
- Yanquan Xu
- Clinical Medicine Research CenterXinqiao HospitalArmy Medical UniversityChongqing400037China
| | - Yu Chen
- Department of Medical OncologyChongqing University Cancer HospitalChongqing400030China
| | - Jiangang Zhang
- Department of Medical OncologyChongqing University Cancer HospitalChongqing400030China
| | - Jingchun Wang
- Department of GastroenterologyXinqiao HospitalArmy Medical UniversityChongqing400037China
| | - Shuai Yang
- Department of PathologyThe 958th HospitalSouthwest HospitalArmy Medical UniversityChongqing400037China
| | - Huakan Zhao
- Department of Medical OncologyChongqing University Cancer HospitalChongqing400030China
| | - Lei Wu
- Department of Medical OncologyChongqing University Cancer HospitalChongqing400030China
| | - Juan Lei
- Department of Medical OncologyChongqing University Cancer HospitalChongqing400030China
| | - Yu Zhou
- Department of Medical OncologyChongqing University Cancer HospitalChongqing400030China
| | - Jin Peng
- Clinical Medicine Research CenterXinqiao HospitalArmy Medical UniversityChongqing400037China
| | - Lu Jiang
- Clinical Medicine Research CenterXinqiao HospitalArmy Medical UniversityChongqing400037China
| | - Qian Chen
- Clinical Medicine Research CenterXinqiao HospitalArmy Medical UniversityChongqing400037China
| | - Rong Xin
- Clinical Medicine Research CenterXinqiao HospitalArmy Medical UniversityChongqing400037China
| | - Jianyun Zhou
- Clinical Medicine Research CenterXinqiao HospitalArmy Medical UniversityChongqing400037China
- Department of GastroenterologyXinqiao HospitalArmy Medical UniversityChongqing400037China
| | - Yongsheng Li
- Clinical Medicine Research CenterXinqiao HospitalArmy Medical UniversityChongqing400037China
- Department of Medical OncologyChongqing University Cancer HospitalChongqing400030China
| |
Collapse
|
26
|
Tang Y, Zhang Y, Chen C, Cao Y, Wang Q, Tang C. Gut microbiota: A new window for the prevention and treatment of neuropsychiatric disease. J Cent Nerv Syst Dis 2025; 17:11795735251322450. [PMID: 39989718 PMCID: PMC11846125 DOI: 10.1177/11795735251322450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/11/2024] [Accepted: 01/27/2025] [Indexed: 02/25/2025] Open
Abstract
Under normal physiological conditions, gut microbiota and host mutually coexist. They play key roles in maintaining intestinal barrier integrity, absorption, and metabolism, as well as promoting the development of the central nervous system (CNS) and emotional regulation. The dysregulation of gut microbiota homeostasis has attracted significant research interest, specifically in its impact on neurological and psychiatric disorders. Recent studies have highlighted the important role of the gut- brain axis in conditions including Alzheimer's Disease (AD), Parkinson's Disease (PD), and depression. This review aims to elucidate the regulatory mechanisms by which gut microbiota affect the progression of CNS disorders via the gut-brain axis. Additionally, we discuss the current research landscape, identify gaps, and propose future directions for microbial interventions against these diseases. Finally, we provide a theoretical reference for clinical treatment strategies and drug development for AD, PD, and depression.
Collapse
Affiliation(s)
- Yali Tang
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yizhu Zhang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Chen Chen
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Ying Cao
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People’s Republic of China
| | - Qiaona Wang
- School of Ecology and Applied Meteorology, Nanjing University of Information Science & Technology, Nanjing, People’s Republic of China
| | - Chuanfeng Tang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
27
|
Jirakran K, Almulla AF, Jaipinta T, Vasupanrajit A, Jansem P, Tunvirachaisakul C, Dzhambazova E, Stoyanov DS, Maes M. Increased atherogenicity in mood disorders: a systematic review, meta-analysis and meta-regression. Neurosci Biobehav Rev 2025; 169:106005. [PMID: 39793682 DOI: 10.1016/j.neubiorev.2025.106005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/12/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) and bipolar disorder (BD) often coexist with metabolic syndrome. Both are linked to increased atherogenicity and a higher risk of cardiovascular diseases. Nevertheless, a comprehensive analysis of key atherogenic biomarkers in MDD/BD is still lacking. OBJECTIVES This meta-analysis evaluates the relationship between atherogenic indices and MDD/BD, while identifying the most effective atherogenic biomarker. METHODS This study adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. We searched electronic databases, including PubMed, Google Scholar, and Web of Science, for articles published up to August 1, 2024. RESULTS We included 85 eligible studies (14 on BD and 71 on MDD), covering 70,856 participants: 18,738 patients and 52,118 healthy controls. MDD/BD patients showed significant increases (p < 0.001) in the Castelli Risk Index 2 (CRI2), Atherogenic Index of Plasma (AIP), and (triglyceride or TG + low-density lipoprotein + very low-density lipoprotein)/(high-density lipoprotein cholesterol or HDL + Apolipoprotein A or ApoA) ratio, but not CRI1 and ApoB/ApoA ratio. Significant lower HDL and lecithin: cholesterol acyltransferase activity, and higher TG levels were observed in MDD/BD patients compared with controls. There were no significant differences between MDD and BD patients. Most included studies lacked the most essential information on the inclusion and exclusion of important confounders. CONCLUSIONS AIP is the most effective atherogenicity index for mood disorders. Regular lipid profiling and metabolic syndrome screening are crucial in MDD/BD. Early intervention with lipid-lowering therapies is recommended to prevent the worsening of atherogenicity and disease progression.
Collapse
Affiliation(s)
- Ketsupar Jirakran
- PhD Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand; Center of Excellence for Maximizing Children's Developmental Potential, Department of Pediatric, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Abbas F Almulla
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand; Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China; Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Thapanee Jaipinta
- PhD Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand.
| | - Asara Vasupanrajit
- PhD Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand.
| | - Priabprat Jansem
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand.
| | - Chavit Tunvirachaisakul
- PhD Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Drozdstoj St Stoyanov
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University Plovdiv, Plovdiv, Bulgaria; Research and Innovation Program for the Development of MU - PLOVDIV- (SRIPD-MUP), Creation of a network of research higher schools, National plan for recovery and sustainability, European Union - NextGenerationEU, Bulgaria
| | - Michael Maes
- PhD Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand; Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, China; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University Plovdiv, Plovdiv, Bulgaria; Research and Innovation Program for the Development of MU - PLOVDIV- (SRIPD-MUP), Creation of a network of research higher schools, National plan for recovery and sustainability, European Union - NextGenerationEU, Bulgaria; Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, South Korea.
| |
Collapse
|
28
|
Tiley JB, Beaudoin JJ, Derebail VK, Murphy WA, Park CC, Veeder JA, Tran L, Beers JL, Jia W, Stewart PW, Brouwer KL. Altered bile acid and coproporphyrin-I disposition in patients with autosomal dominant polycystic kidney disease. Br J Clin Pharmacol 2025; 91:353-364. [PMID: 39317666 PMCID: PMC12001807 DOI: 10.1111/bcp.16221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 06/22/2024] [Accepted: 07/30/2024] [Indexed: 09/26/2024] Open
Abstract
AIMS Serum, liver and urinary bile acids are increased, and hepatic transport protein levels are decreased in a non-clinical model of polycystic kidney disease. Similar changes in patients with autosomal dominant polycystic kidney disease (ADPKD) may predispose them to drug-induced liver injury (DILI) and hepatic drug-drug interactions (DDIs). Systemic coproporphyrin-I (CP-I), an endogenous biomarker for hepatic OATP1B function and MRP2 substrate, is used to evaluate OATP1B-mediated DDI risk in humans. In this clinical observational cohort-comparison study, bile acid profiles and CP-I concentrations in healthy volunteers and patients with ADPKD were compared. METHODS Serum and urine samples from healthy volunteers (n = 16) and patients with ADPKD (n = 8) were collected. Serum bile acids, and serum and urine CP-I concentrations, were quantified by ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). RESULTS Patients with ADPKD exhibited increased serum concentrations of total (1.3-fold) and taurine-conjugated (2.8-fold) bile acids compared to healthy volunteers. Specifically, serum concentrations of six bile acids known to be more hydrophobic/hepatotoxic (glycochenodeoxycholate, taurochenodeoxycholate, taurodeoxycholate, lithocholate, glycolithocholate and taurolithocholate) were increased (1.5-, 2.9-, 2.8-, 1.6-, 1.7- and 2.7-fold, respectively) in patients with ADPKD. Furthermore, serum CP-I concentrations were elevated and the renal clearance of CP-I was reduced in patients with ADPKD compared to healthy volunteers. CONCLUSIONS Increased exposure to bile acids may increase susceptibility to DILI in some patients with ADPKD. Furthermore, the observed increase in serum CP-I concentrations could be attributed, in part, to impaired OATP1B function in patients with ADPKD, which could increase the risk of DDIs involving OATP1B substrates compared to healthy volunteers.
Collapse
Affiliation(s)
- Jacqueline B. Tiley
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - James J. Beaudoin
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Vimal K. Derebail
- UNC Kidney Center, Division of Nephrology and Hypertension, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - William A. Murphy
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Christine C. Park
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Justin A. Veeder
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lana Tran
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jessica L. Beers
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wei Jia
- University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Paul W. Stewart
- Department of Biostatistics, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kim L.R. Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
29
|
Won TH, Arifuzzaman M, Parkhurst CN, Miranda IC, Zhang B, Hu E, Kashyap S, Letourneau J, Jin WB, Fu Y, Guzior DV, Quinn RA, Guo CJ, David LA, Artis D, Schroeder FC. Host metabolism balances microbial regulation of bile acid signalling. Nature 2025; 638:216-224. [PMID: 39779854 PMCID: PMC11886927 DOI: 10.1038/s41586-024-08379-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 11/08/2024] [Indexed: 01/11/2025]
Abstract
Metabolites derived from the intestinal microbiota, including bile acids (BA), extensively modulate vertebrate physiology, including development1, metabolism2-4, immune responses5-7 and cognitive function8. However, to what extent host responses balance the physiological effects of microbiota-derived metabolites remains unclear9,10. Here, using untargeted metabolomics of mouse tissues, we identified a family of BA-methylcysteamine (BA-MCY) conjugates that are abundant in the intestine and dependent on vanin 1 (VNN1), a pantetheinase highly expressed in intestinal tissues. This host-dependent MCY conjugation inverts BA function in the hepatobiliary system. Whereas microbiota-derived free BAs function as agonists of the farnesoid X receptor (FXR) and negatively regulate BA production, BA-MCYs act as potent antagonists of FXR and promote expression of BA biosynthesis genes in vivo. Supplementation with stable-isotope-labelled BA-MCY increased BA production in an FXR-dependent manner, and BA-MCY supplementation in a mouse model of hypercholesteraemia decreased lipid accumulation in the liver, consistent with BA-MCYs acting as intestinal FXR antagonists. The levels of BA-MCY were reduced in microbiota-deficient mice and restored by transplantation of human faecal microbiota. Dietary intervention with inulin fibre further increased levels of both free BAs and BA-MCY levels, indicating that BA-MCY production by the host is regulated by levels of microbiota-derived free BAs. We further show that diverse BA-MCYs are also present in human serum. Together, our results indicate that BA-MCY conjugation by the host balances host-dependent and microbiota-dependent metabolic pathways that regulate FXR-dependent physiology.
Collapse
Affiliation(s)
- Tae Hyung Won
- Department of Chemistry and Chemical Biology, Boyce Thompson Institute, Cornell University, Ithaca, NY, USA
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon-si, Republic of Korea
| | - Mohammad Arifuzzaman
- Joan and Sanford I. Weill Department of Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Christopher N Parkhurst
- Joan and Sanford I. Weill Department of Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Isabella C Miranda
- Joan and Sanford I. Weill Department of Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Bingsen Zhang
- Department of Chemistry and Chemical Biology, Boyce Thompson Institute, Cornell University, Ithaca, NY, USA
| | - Elin Hu
- Joan and Sanford I. Weill Department of Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Sanchita Kashyap
- Joan and Sanford I. Weill Department of Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jeffrey Letourneau
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Wen-Bing Jin
- Joan and Sanford I. Weill Department of Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Yousi Fu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Douglas V Guzior
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI, USA
| | - Robert A Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Chun-Jun Guo
- Joan and Sanford I. Weill Department of Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Lawrence A David
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
- Program in Computational Biology and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - David Artis
- Joan and Sanford I. Weill Department of Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Allen Discovery Center for Neuroimmune Interactions, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| | - Frank C Schroeder
- Department of Chemistry and Chemical Biology, Boyce Thompson Institute, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
30
|
Wang P, Ouyang H, Bi G, Liang F, Hu S, Wu C, Jiang X, Zhou W, Li D, Zhang S, Yang X, Zhao M, Fang JH, Wang H, Jia W, Zhu ZJ, Bi H. Schisandrol B alleviates depression-like behavior in mice by regulating bile acid homeostasis in the brain-liver-gut axis via the pregnane X receptor. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156340. [PMID: 39809031 DOI: 10.1016/j.phymed.2024.156340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/18/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Depression is a widely recognized neuropsychiatric disorder. Recent studies have shown a potential correlation between bile acid disorders and depression, highlighting the importance of maintaining bile acid balance for effective antidepressant treatment. Schisandrol B (SolB), a primary bioactive compound from Schisandra chinensis (Turcz.) Baill. or Schisandra sphenanthera Rehd.etWils, is pivotal in regulating bile acid homeostasis via pregnane X receptor (PXR) in cholestasis. However, the potential of SolB in alleviating depression-like symptoms, its pharmacological effects, and the underlying mechanisms remain to be fully elucidated. METHODS We confirmed the effect of SolB against depression induced by chronic restraint stress (CRS) and chronic unpredictable mild stress (CUMS) in mice. The role of SolB in bile acid homeostasis in depression was analyzed using the metabolomic. Gene analyses and 16S rRNA sequencing were employed to investigate the involvement of PXR. Experiments with Pxr-/- mice were conducted to confirm the essential role of the PXR pathway in SolB's antidepressant effects. RESULTS SolB treatment significantly increased sucrose consumption in the SPT and the locomotor activity in the OFT, while decreasing immobility time in the FST and TST in mice exposed to CRS and CUMS. Additionally, SolB treatment significantly preserved the integrity of the dendritic spine, elevated synaptic protein PSD95 levels, and augmented CREB/BDNF expression. Metabolomic and gene analyses indicated that SolB treatment significantly facilitated bile acid metabolism, promoted intestinal bile acid efflux, decreased hippocampal levels of the secondary bile acids DCA and TLCA, and upregulated expression of the PXR target proteins CYP3A11, SULT2A1, MRP2, and OATP1B1 in the liver, and MRP2 and MDR1 in hippocampus, which are integral to bile acid homeostasis. 16S rRNA sequencing revealed that SolB reduced the abundance of the bile salt hydrolase (BSH)-producing bacteria Lactobacillus johnsonii and Bacteroides fragilis and subsequently decreased the production of TLCA and DCA. Moreover, SolB failed to protect against depression induced by CRS in Pxr-null mice, suggesting that the antidepressant effect of SolB was PXR-dependent. CONCLUSIONS These results provide direct evidence of the antidepressant effect of SolB via activation of PXR to regulate bile acid homeostasis in the brain-liver-gut axis, suggesting that SolB may serve as a novel potential target for preventing and treating depression.
Collapse
Affiliation(s)
- Peng Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Hui Ouyang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Guofang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Fengting Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuang Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Chenghua Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaowen Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wenhong Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Dan Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuaishuai Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China; The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, China
| | - Mingliang Zhao
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jian-Hong Fang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Haitao Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wei Jia
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.
| | - Huichang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China; The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, China.
| |
Collapse
|
31
|
Ferraz ÁAB, Vianna CFM, Henriques DF, Gorgulho GCF, Santa-Cruz F, Siqueira LT, Kreimer F. The Impact of Cholecystectomy on the Metabolic Profile of Patients Previously Submitted to Bariatric Surgery. Surg Laparosc Endosc Percutan Tech 2025; 35:e1348. [PMID: 39618187 DOI: 10.1097/sle.0000000000001348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE To evaluate the influence of late cholecystectomy following bariatric surgery on the postoperative evolution of weight loss and biochemical, metabolic, and micronutrient parameters. METHODS A retrospective study that assessed 86 patients who underwent cholecystectomy after at least 18 months of bariatric surgery. The analyzed variables included demographic data, comorbidities, weight loss, and biochemical, metabolic, and micronutrient parameters. RESULTS Among the analyzed patients, 20 underwent gastric bypass (GB) and 66 underwent sleeve gastrectomy (SG). The GB group comprised 55% of women, with a mean age of 54.4 years and a mean preoperative body mass index (BMI) of 29.2 kg/m 2 . The mean time elapsed between GB and cholecystectomy was 118.3±43.9 months. The sample of SG comprised 83.3% of women, with a mean age of 41.1 years and a mean preoperative BMI of 28.7 kg/m 2 . The mean time elapsed between SG and cholecystectomy was 26.1±17.5 months. Both SG and GB groups showed a reduction in the mean BMI, but it was not statistically significant after cholecystectomy. In the metabolic, biochemical, and micronutrient evaluation, there was no statistically significant difference, except in the GB group, where an increase in vitamin D was observed after cholecystectomy with statistical significance. CONCLUSION Cholecystectomy does not negatively impact the clinical and anthropometric evolution of patients previously submitted to bariatric surgery.
Collapse
Affiliation(s)
| | - Cassio F M Vianna
- Medical School, Federal University of Pernambuco, Recife, PE, Brazil
| | | | | | | | | | | |
Collapse
|
32
|
Zhu Y, Zhang KX, Bu QY, Song SX, Chen Y, Zou H, You XY, Zhao GP. Ginsenosides From Panax ginseng Improves Hepatic Lipid Metabolism Disorders in HFD-Fed Rats by Regulating Gut Microbiota and Cholesterol Metabolism Signaling Pathways. Phytother Res 2025; 39:714-732. [PMID: 39660634 DOI: 10.1002/ptr.8402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/11/2024] [Accepted: 02/27/2024] [Indexed: 12/12/2024]
Abstract
A high-fat diet (HFD) is often associated with hepatic lipid metabolism disorders, leading to dysfunction in multiple body systems. Ginsenosides derived from Panax ginseng have been reported to possess potential effects in ameliorating lipid metabolism disorders; however, their underlying mechanisms remain insufficiently explored. This study aims to investigate the bioactivities of ginsenosides in combating lipid metabolism disorders and obesity, with a focus on their mechanisms involving the cholesterol metabolism signaling pathway and gut microbiota. Our results demonstrated that ginsenoside treatment significantly reduced overall body weight, body weight changes, liver weight, and eWAT weight, as well as alleviated hepatic steatosis and dyslipidemia in HFD-fed rats, without affecting food intake. These effects were dose-dependent. Furthermore, 16S rRNA sequencing revealed that ginsenosides significantly increased the relative abundance of Akkermansia muciniphila, Blautia, Eisenbergiella, Clostridium clusters XI, XVIII, and III, while decreasing the relative abundance of Clostridium subcluster XIVa and Dorea. In addition, ginsenoside treatment significantly regulated the expression of hepatic genes and proteins involved in the cholesterol metabolism signaling pathway (FXR, CYP7A1, CYP7B1, CYP27A1, ABCG5, ABCG8, Insig2, and Dhcr7), potentially inhibiting hepatic cholesterol biosynthesis while promoting cholesterol transport to HDL and its excretion via bile and feces. Notably, levels of 7-dehydrocholesterol (7-DHC) and 27-hydroxycholesterol (27-OHC) were reduced, while 5β,6β-epoxycholesterol (5,6β-epoxy) levels were elevated following ginsenoside treatment, indicating significant modulation of oxysterols by ginsenosides. Moreover, bile acid enterohepatic circulation was regulated through the enhancement of hepatic FXR-CYP7A1 signaling and intestinal FXR-FGF15 signaling in HFD-fed rats treated with ginsenosides, which was closely linked to gut microbiota composition. Collectively, our findings suggest that ginsenosides alleviate hepatic lipid metabolism disorders by modulating gut microbiota and the cholesterol metabolism signaling pathway in HFD-fed rats.
Collapse
Affiliation(s)
- Yue Zhu
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Kang-Xi Zhang
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Qing-Yun Bu
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- Haihe Laboratory of Synthetic Biology, Tianjin, China
| | - Shu-Xia Song
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- Haihe Laboratory of Synthetic Biology, Tianjin, China
| | - Yue Chen
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Hong Zou
- CAS Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Xiao-Yan You
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- Henan Engineering Research Center of Food Microbiology, College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Guo-Ping Zhao
- Master Lab for Innovative Application of Nature Products, National Center of Technology Innovation for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
33
|
Yang Q, Luo Q, Xia W, Yao N, Wang F, Xie C, Zhang H, He Y. Study on the mechanism on Yi-guan-jian decoction alleviating cognitive dysfunction in type 2 diabetes mellitus. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119286. [PMID: 39725366 DOI: 10.1016/j.jep.2024.119286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yi-guan-jian decoction (YGJ) is a traditional Chinese medicine prescription commonly used for treating syndromes associated with Yin deficiency in the liver and kidney, as well as Qi-obstructed in liver. AIM OF THE STUDY YGJ has shown potential alleviating cognitive dysfunction in type 2 diabetes mellitus (T2DM). However, the precise mechanisms are not yet fully understood. This study aims to reveal the mechanism by which YGJ alleviates cognitive dysfunction in T2DM. MATERIALS AND METHODS Various doses of YGJ were administered to T2DM rats with cognitive dysfunction for 8 weeks. The positive control group received a combination of metformin and memantine. Cognitive function was assessed in T2DM rats using the Morris water maze test during treatment. Changes in gut microbiota and bile acids in the intestine were evaluated, and their interactions analyzed. Additionally, this study also evaluated the expressions of inflammatory markers (IL-1β,TNF-α, IL-16, IL-18 and CRP protein), Tau protein, neurotransmitter (5-HT and GABA), and bile acid receptor (FXR, PXR, VDR, and TGR5). RESULTS YGJ significantly alleviated insulin resistance and hyperlipidemia, reduce the levels of inflammatory factors in serum and hippocampus, and decreased mortality in T2DM rats. The Morris water maze test indicated that YGJ reduced the escape latency and increased platform crossing frequency, thereby improving cognitive function in T2DM rats. Furthermore, YGJ regulated the abundance of microorganisms associated with bile acid metabolism, including Romboutsia, Bacteroides, Turicibacter, Blautia, and Ruminococcus, thus regulating bile acid metabolism in T2DM rats. Additionally, YGJ also regulated bile acid metabolism by regulating intestinal FXR, PXR, VDR and TRG5 receptors. CONCLUSION YGJ can alleviate glucose homeostasis, insulin sensitivity, lipid metabolism, neuroinflammation, cognitive function, as well as remodel intestinal flora and BA composition in CDT2DM rats, which is a potential complementary and alternative therapy for the prevention and treatment of CDT2DM. These effects may be associated that YGJ regulates the structure of intestinal flora and BA metabolism, and inhibits intestinal BA receptors FXR, PXR, TGR5, and VDR.
Collapse
Affiliation(s)
- Qiyue Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China.
| | - Qiwei Luo
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China.
| | - Wenrui Xia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China.
| | - Nairong Yao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China.
| | - Fang Wang
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China.
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, PR China.
| | - Haiyan Zhang
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, PR China.
| | - Yanan He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| |
Collapse
|
34
|
Ma X, Yu J, Ma Y, Huang X, Zhu K, Jiang Z, Zhang L, Liu Y. Explore the mechanism of yishenjiangya formula in the treatment of senile hypertension based on multi-omics technology. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118886. [PMID: 39362324 DOI: 10.1016/j.jep.2024.118886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Yishenjiangya formula (YSJ) is a traditional Chinese medicine (TCM) primarily composed of qi-tonifying components. This classic formula is commonly utilized to treat kidney qi deficiency in elderly patients with hypertension. According to TCM, maintaining a balance between qi and blood is crucial for stable blood pressure. Kidney qi deficiency can disrupt this balance, altering fluid shear force and, ultimately, leading to hypertension, particularly in elderly populations. Despite YSJ's efficacy in treating hypertension, its specific anti-hypertensive mechanisms remain unclear. AIM OF THE STUDY YSJ is commonly prescribed for elderly patients with hypertension. Earlier metabolomics studies demonstrated that YSJ exerts antihypertensive effects by influencing four key pathways: linoleic acid metabolism, glycerol phospholipid metabolism, arginine and proline metabolism, and steroid hormone biosynthesis. This study aims to combine metabolomic and proteomic analyses to thoroughly understand the molecular biological mechanisms responsible for YSJ's anti-hypertensive properties. METHODS Ultra-Performance Liquid Chromatography-Mass Spectrometry (UPLC-MS) metabolomics, combined with Label-Free Quantitation (LFQ) proteomics, was employed to analyze serum samples from elderly individuals with and without hypertension pre- and post-YSJ intervention. Serum levels of candidate proteins were assessed using enzyme-linked immunosorbent assay, and receiver operating characteristic curves were used to evaluate the diagnostic performance of the target proteins. RESULTS Eight differentially expressed metabolites and three differentially expressed proteins were identified as potential therapeutic targets of YSJ. These substances are primarily involved in unsaturated fatty acid metabolism, fluid shear stress and atherosclerosis pathway, primary bile acid biosynthesis, proline metabolism, apoptosis, and endoplasmic reticulum stress. YSJ exerts its therapeutic effects on hypertension in the elderly by modulating these pathways. CONCLUSIONS YSJ effectively treats senile hypertension. By analyzing the correlation between therapeutic targets and pathways, YSJ's anti-hypertensive effect was achieved by inhibiting lipid peroxidation and matrix degeneration. Combining metabolomics and proteomics provides an effective method for uncovering YSJ's anti-hypertensive mechanisms.
Collapse
Affiliation(s)
- Xu Ma
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Jie Yu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, Shandong, China
| | - Yongbo Ma
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, Shanghai, China
| | - Xinyu Huang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Kunpeng Zhu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Zhen Jiang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Lei Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, Shandong, China.
| | - Yingying Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, Shandong, China.
| |
Collapse
|
35
|
Liu Z, You C. The bile acid profile. Clin Chim Acta 2025; 565:120004. [PMID: 39419312 DOI: 10.1016/j.cca.2024.120004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
As a large and structurally diverse family of small molecules, bile acids play a crucial role in regulating lipid, glucose, and energy metabolism. In the human body, bile acids share a similar chemical structure with many isomers, exhibit little difference in polarity, and possess various physiological activities. The types and contents of bile acids present in different diseases vary significantly. Therefore, comprehensive and accurate detection of the content of various types of bile acids in different biological samples can not only provide new insights into the pathogenesis of diseases but also facilitate the exploration of novel strategies for disease diagnosis, treatment, and prognosis. The detection of disease-induced changes in bile acid profiles has emerged as a prominent research focus in recent years. Concurrently, targeted metabolomics methods utilizing high-performance liquid chromatography-mass spectrometry (HPLC-MS) have progressively established themselves as the predominant technology for the separation and detection of bile acids. Bile acid profiles will increasingly play an important role in diagnosis and guidance in the future as the relationship between disease and changes in bile acid profiles becomes clearer. This highlights the growing diagnostic value of bile acid profiles and their potential to guide clinical decision-making. This review aims to explore the significance of bile acid profiles in clinical diagnosis from four perspectives: the synthesis and metabolism of bile acids, techniques for detecting bile acid profiles, changes in bile acid profiles associated with diseases, and the challenges and future prospects of applying bile acid profiles in clinical settings.
Collapse
Affiliation(s)
- Zhenhua Liu
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Chongge You
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
| |
Collapse
|
36
|
Du J, Shen M, Chen J, Yan H, Xu Z, Yang X, Yang B, Luo P, Ding K, Hu Y, He Q. The impact of solute carrier proteins on disrupting substance regulation in metabolic disorders: insights and clinical applications. Front Pharmacol 2025; 15:1510080. [PMID: 39850557 PMCID: PMC11754210 DOI: 10.3389/fphar.2024.1510080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025] Open
Abstract
Carbohydrates, lipids, bile acids, various inorganic salt ions and organic acids are the main nutrients or indispensable components of the human body. Dysregulation in the processes of absorption, transport, metabolism, and excretion of these metabolites can lead to the onset of severe metabolic disorders, such as type 2 diabetes, non-alcoholic fatty liver disease, gout and hyperbilirubinemia. As the second largest membrane receptor supergroup, several major families in the solute carrier (SLC) supergroup have been found to play key roles in the transport of substances such as carbohydrates, lipids, urate, bile acids, monocarboxylates and zinc ions. Based on common metabolic dysregulation and related metabolic substances, we explored the relationship between several major families of SLC supergroup and metabolic diseases, providing examples of drugs targeting SLC proteins that have been approved or are currently in clinical/preclinical research as well as SLC-related diagnostic techniques that are in clinical use or under investigation. By highlighting these connections, we aim to provide insights that may contribute to the development of improved treatment strategies and targeted therapies for metabolic disorders.
Collapse
Affiliation(s)
- Jiangxia Du
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Minhui Shen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiajia Chen
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kefeng Ding
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuhuai Hu
- Yuhong Pharmaceutical Technology Co., Ltd., Hangzhou, Zhejiang, China
| | - Qiaojun He
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Department of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
37
|
Enejoh OA, Okonkwo CH, Nortey H, Kemiki OA, Moses A, Mbaoji FN, Yusuf AS, Awe OI. Machine learning and molecular dynamics simulations predict potential TGR5 agonists for type 2 diabetes treatment. Front Chem 2025; 12:1503593. [PMID: 39850718 PMCID: PMC11754275 DOI: 10.3389/fchem.2024.1503593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/13/2024] [Indexed: 01/25/2025] Open
Abstract
Introduction Treatment of type 2 diabetes (T2D) remains a significant challenge because of its multifactorial nature and complex metabolic pathways. There is growing interest in finding new therapeutic targets that could lead to safer and more effective treatment options. Takeda G protein-coupled receptor 5 (TGR5) is a promising antidiabetic target that plays a key role in metabolic regulation, especially in glucose homeostasis and energy expenditure. TGR5 agonists are attractive candidates for T2D therapy because of their ability to improve glycemic control. This study used machine learning-based models (ML), molecular docking (MD), and molecular dynamics simulations (MDS) to explore novel small molecules as potential TGR5 agonists. Methods Bioactivity data for known TGR5 agonists were obtained from the ChEMBL database. The dataset was cleaned and molecular descriptors based on Lipinski's rule of five were selected as input features for the ML model, which was built using the Random Forest algorithm. The optimized ML model was used to screen the COCONUT database and predict potential TGR5 agonists based on their molecular features. 6,656 compounds predicted from the COCONUT database were docked within the active site of TGR5 to calculate their binding energies. The four top-scoring compounds with the lowest binding energies were selected and their activities were compared to those of the co-crystallized ligand. A 100 ns MDS was used to assess the binding stability of the compounds to TGR5. Results Molecular docking results showed that the lead compounds had a stronger affinity for TGR5 than the cocrystallized ligand. MDS revealed that the lead compounds were stable within the TGR5 binding pocket. Discussion The combination of ML, MD, and MDS provides a powerful approach for predicting new TGR5 agonists that can be optimised for T2D treatment.
Collapse
Affiliation(s)
- Ojochenemi A. Enejoh
- Genetics, Genomics and Bioinformatics Department, National Biotechnology Research and Development Agency, Abuja, Nigeria
| | | | - Hector Nortey
- Department of Clinical Pathology, Noguchi Memorial Institute for Medical Research, College of Health Science, University of Ghana, Accra, Ghana
| | - Olalekan A. Kemiki
- Molecular and Tissue Culture Laboratory, Babcock University, Ilisan-remo, Ogun State, Nigeria
| | - Ainembabazi Moses
- African Centers of Excellence in Bioinformatics and data intensive sciences, Department of Immunology and Microbiology, Makerere University, Makerere, Uganda
- Infectious Disease Institute (IDI), Makerere University, Kampala, Uganda
| | - Florence N. Mbaoji
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Enugu, Nigeria
| | - Abdulrazak S. Yusuf
- Department of Biochemistry, Faculty of Basic Health Science, Bayero University, Kano, Nigeria
| | - Olaitan I. Awe
- African Society for Bioinformatics and Computational Biology, Cape Town, South Africa
| |
Collapse
|
38
|
Chen WY, Zhang JH, Chen LL, Byrne CD, Targher G, Luo L, Ni Y, Zheng MH, Sun DQ. Bioactive metabolites: A clue to the link between MASLD and CKD? Clin Mol Hepatol 2025; 31:56-73. [PMID: 39428978 PMCID: PMC11791555 DOI: 10.3350/cmh.2024.0782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 10/22/2024] Open
Abstract
Metabolites produced as intermediaries or end-products of microbial metabolism provide crucial signals for health and diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD). These metabolites include products of the bacterial metabolism of dietary substrates, modification of host molecules (such as bile acids [BAs], trimethylamine-N-oxide, and short-chain fatty acids), or products directly derived from bacteria. Recent studies have provided new insights into the association between MASLD and the risk of developing chronic kidney disease (CKD). Furthermore, alterations in microbiota composition and metabolite profiles, notably altered BAs, have been described in studies investigating the association between MASLD and the risk of CKD. This narrative review discusses alterations of specific classes of metabolites, BAs, fructose, vitamin D, and microbiota composition that may be implicated in the link between MASLD and CKD.
Collapse
Affiliation(s)
- Wen-Ying Chen
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jia-Hui Zhang
- Department of Pediatric Laboratory, Affiliated Children’s Hospital of Jiangnan University, Wuxi Children’s Hospital, Wuxi, Jiangsu, China
| | - Li-Li Chen
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Christopher D. Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton General Hospital, Southampton, UK
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore - Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Liang Luo
- Intensive Care Medicine, Jiangnan University Medical Center, Wuxi, China
| | - Yan Ni
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Dan-Qin Sun
- Urologic Nephrology Center, Jiangnan University Medical Center, Wuxi, China
- Affiliated Wuxi Clinical College of Nantong University, Wuxi, China
- Department of Nephrology, Wuxi No.2 People’s Hospital, Wuxi, China
| |
Collapse
|
39
|
Kastrinou-Lampou V, Rodríguez-Pérez R, Poller B, Huth F, Schadt HS, Kullak-Ublick GA, Arand M, Camenisch G. Drug-induced cholestasis (DIC) predictions based on in vitro inhibition of major bile acid clearance mechanisms. Arch Toxicol 2025; 99:377-391. [PMID: 39542928 DOI: 10.1007/s00204-024-03895-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/17/2024] [Indexed: 11/17/2024]
Abstract
Drug-induced cholestasis (DIC) is recognized as a major safety concern in drug development, as it represents one of the three types of drug-induced liver injury (DILI). Cholestasis is characterized by the disruption of bile flow, leading to intrahepatic accumulation of toxic bile acids. Bile acid regulation is a multifarious process, orchestrated by several hepatic mechanisms, namely sinusoidal uptake and efflux, canalicular secretion and intracellular metabolism. In the present study, we developed a prediction model of DIC using in vitro inhibition data for 47 marketed drugs on nine transporters and five enzymes known to regulate bile acid homeostasis. The resulting model was able to distinguish between drugs with or without DILI concern (p-value = 0.039) and demonstrated a satisfactory predictive performance, with the area under the precision-recall curve (PR AUC) measured at 0.91. Furthermore, we simplified the model considering only two processes, namely reversible inhibition of OATP1B1 and time-dependent inhibition of CYP3A4, which provided an enhanced performance (PR AUC = 0.95). Our study supports literature findings suggesting a contribution not only from a single process inhibition, but a rather synergistic effect of the key bile acid clearance processes in the development of cholestasis. The use of a quantitative model in the preclinical investigations of DIC is expected to reduce attrition rate in advanced development programs and guide the discovery and development of safe medicines.
Collapse
Affiliation(s)
- Vlasia Kastrinou-Lampou
- Pharmacokinetic Sciences, BioMedical Research, Novartis, Basel, Switzerland
- Preclinical Safety, BioMedical Research, Novartis, Basel, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Birk Poller
- Pharmacokinetic Sciences, BioMedical Research, Novartis, Basel, Switzerland
| | - Felix Huth
- Pharmacokinetic Sciences, BioMedical Research, Novartis, Basel, Switzerland
| | - Heiko S Schadt
- Preclinical Safety, BioMedical Research, Novartis, Basel, Switzerland
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Mechanistic Safety, CMO and Patient Safety, Global Drug Development, Novartis, Basel, Switzerland
| | - Michael Arand
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Gian Camenisch
- Pharmacokinetic Sciences, BioMedical Research, Novartis, Basel, Switzerland.
| |
Collapse
|
40
|
Fuchs CD, Simbrunner B, Baumgartner M, Campbell C, Reiberger T, Trauner M. Bile acid metabolism and signalling in liver disease. J Hepatol 2025; 82:134-153. [PMID: 39349254 DOI: 10.1016/j.jhep.2024.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 10/02/2024]
Abstract
Bile acids (BAs) serve as signalling molecules, efficiently regulating their own metabolism and transport, as well as key aspects of lipid and glucose homeostasis. BAs shape the gut microbial flora and conversely are metabolised by microbiota. Disruption of BA transport, metabolism and physiological signalling functions contribute to the pathogenesis and progression of a wide range of liver diseases including cholestatic disorders and MASLD (metabolic dysfunction-associated steatotic liver disease), as well as hepatocellular and cholangiocellular carcinoma. Additionally, impaired BA signalling may also affect the intestine and kidney, thereby contributing to failure of gut integrity and driving the progression and complications of portal hypertension, cholemic nephropathy and the development of extrahepatic malignancies such as colorectal cancer. In this review, we will summarise recent advances in the understanding of BA signalling, metabolism and transport, focusing on transcriptional regulation and novel BA-focused therapeutic strategies for cholestatic and metabolic liver diseases.
Collapse
Affiliation(s)
- Claudia D Fuchs
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Benedikt Simbrunner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Maximillian Baumgartner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Clarissa Campbell
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
41
|
Darmanto AG, Yen TL, Jan JS, Linh TTD, Taliyan R, Yang CH, Sheu JR. Beyond metabolic messengers: Bile acids and TGR5 as pharmacotherapeutic intervention for psychiatric disorders. Pharmacol Res 2025; 211:107564. [PMID: 39733841 DOI: 10.1016/j.phrs.2024.107564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Psychiatric disorders pose a significant global health challenge, exacerbated by the COVID-19 pandemic and insufficiently addressed by the current treatments. This review explores the emerging role of bile acids and the TGR5 receptor in the pathophysiology of psychiatric conditions, emphasizing their signaling within the gut-brain axis. We detail the synthesis and systemic functions of bile acids, their transformation by gut microbiota, and their impact across various neuropsychiatric disorders, including major depressive disorder, general anxiety disorder, schizophrenia, autism spectrum disorder, and bipolar disorder. The review highlights how dysbiosis and altered bile acid metabolism contribute to the development and exacerbation of these neuropsychiatric disorders through mechanisms involving inflammation, oxidative stress, and neurotransmitter dysregulation. Importantly, we detail both pharmacological and non-pharmacological interventions that modulate TGR5 signaling, offering potential breakthroughs in treatment strategies. These include dietary adjustments to enhance beneficial bile acids production and the use of specific TGR5 agonists that have shown promise in preclinical and clinical settings for their regulatory effects on critical pathways such as cAMP-PKA, NRF2-mediated antioxidant responses, and neuroinflammation. By integrating findings from the dynamics of gut microbiota, bile acids metabolism, and TGR5 receptor related signaling events, this review underscores cutting-edge therapeutic approaches poised to revolutionize the management and treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Arief Gunawan Darmanto
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; School of Medicine, Universitas Ciputra, Surabaya 60219, Indonesia
| | - Ting-Lin Yen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; Department of Medical Research, Cathay General Hospital, Taipei 22174, Taiwan, ROC
| | - Jing-Shiun Jan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC
| | - Tran Thanh Duy Linh
- Family Medicine Training Center, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Viet Nam
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, Pilani, Rajasthan, India
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; Research Center for Neuroscience, Taipei Medical University, Taipei, Taiwan, ROC.
| | - Joen-Rong Sheu
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; Research Center for Neuroscience, Taipei Medical University, Taipei, Taiwan, ROC; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, ROC.
| |
Collapse
|
42
|
Li W, Gao W, Yan S, Yang L, Zhu Q, Chu H. Gut Microbiota as Emerging Players in the Development of Alcohol-Related Liver Disease. Biomedicines 2024; 13:74. [PMID: 39857657 PMCID: PMC11761646 DOI: 10.3390/biomedicines13010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 12/23/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
The global incidence and mortality rates of alcohol-related liver disease are on the rise, reflecting a growing health concern worldwide. Alcohol-related liver disease develops due to a complex interplay of multiple reasons, including oxidative stress generated during the metabolism of ethanol, immune response activated by immunogenic substances, and subsequent inflammatory processes. Recent research highlights the gut microbiota's significant role in the progression of alcohol-related liver disease. In patients with alcohol-related liver disease, the relative abundance of pathogenic bacteria, including Enterococcus faecalis, increases and is positively correlated with the level of severity exhibited by alcohol-related liver disease. Supplement probiotics like Lactobacillus, as well as Bifidobacterium, have been found to alleviate alcohol-related liver disease. The gut microbiota is speculated to trigger specific signaling pathways, influence metabolite profiles, and modulate immune responses in the gut and liver. This research aimed to investigate the role of gut microorganisms in the onset and advancement of alcohol-related liver disease, as well as to uncover the underlying mechanisms by which the gut microbiota may contribute to its development. This review outlines current treatments for reversing gut dysbiosis, including probiotics, fecal microbiota transplantation, and targeted phage therapy. Particularly, targeted therapy will be a vital aspect of future alcohol-related liver disease treatment. It is to be hoped that this article will prove beneficial for the treatment of alcohol-related liver disease.
Collapse
Affiliation(s)
- Wei Li
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Hubei Clinical Research Center for Infectious Diseases, Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan 430023, China;
| | - Wenkang Gao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.G.); (S.Y.); (L.Y.)
| | - Shengqi Yan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.G.); (S.Y.); (L.Y.)
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.G.); (S.Y.); (L.Y.)
| | - Qingjing Zhu
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Hubei Clinical Research Center for Infectious Diseases, Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan 430023, China;
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.G.); (S.Y.); (L.Y.)
| |
Collapse
|
43
|
Qi Y, Ma Y, Duan G. Pharmacological Mechanisms of Bile Acids Targeting the Farnesoid X Receptor. Int J Mol Sci 2024; 25:13656. [PMID: 39769418 PMCID: PMC11727972 DOI: 10.3390/ijms252413656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025] Open
Abstract
Bile acids (BAs), a category of amphiphilic metabolites synthesized by liver cells and released into the intestine via the bile duct, serve a vital role in the emulsification of ingested fats during the digestive process. Beyond their conventional emulsifying function, BAs, with their diverse structures, also act as significant hormones within the body. They are pivotal in facilitating nutrient absorption by interacting with the farnesoid X receptor (FXR), and they serve as key regulators of lipid and glucose metabolism, as well as immune system balance. Consequently, BAs contribute to the metabolism of glucose and lipids, enhance the digestion and absorption of lipids, and maintain the equilibrium of the bile pool. Their actions are instrumental in addressing obesity, managing cholestasis, and treating diabetes, and are involved in the onset and progression of cancer. This paper presents an updated systematic review of the pharmacological mechanisms by which BAs target the FXR, incorporating recent findings and discussing their signaling pathways in the context of novel research, including their distinct roles in various disease states and populations. The aim is to provide a theoretical foundation for the continued research and clinical application of BAs.
Collapse
Affiliation(s)
- Youchao Qi
- College of Agriculture and Animal Husbandry, Qinghai University, Xining 810016, China;
- Academy of Agriculture and Forestry Sciences, Qinghai University, Xining 810016, China
- Qinghai Plateau Key Laboratory of Tree Genetics and Breeding, Xining 810016, China
- Key Laboratory of Medicinal Animal and Plant Resources of Qinghai Tibetan Plateau, Qinghai Normal University, Xining 810008, China;
- Academy of Plateau Science and Sustainability, Qinghai Normal University, Xining 810008, China
| | - Yonggui Ma
- Key Laboratory of Medicinal Animal and Plant Resources of Qinghai Tibetan Plateau, Qinghai Normal University, Xining 810008, China;
- Academy of Plateau Science and Sustainability, Qinghai Normal University, Xining 810008, China
| | - Guozhen Duan
- Academy of Agriculture and Forestry Sciences, Qinghai University, Xining 810016, China
- Qinghai Plateau Key Laboratory of Tree Genetics and Breeding, Xining 810016, China
| |
Collapse
|
44
|
Sinclair DA. A bile acid could explain how calorie restriction slows ageing. Nature 2024:10.1038/d41586-024-04062-1. [PMID: 39695280 DOI: 10.1038/d41586-024-04062-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
|
45
|
Li Y, Wang L, Yi Q, Luo L, Xiong Y. Regulation of bile acids and their receptor FXR in metabolic diseases. Front Nutr 2024; 11:1447878. [PMID: 39726876 PMCID: PMC11669848 DOI: 10.3389/fnut.2024.1447878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/13/2024] [Indexed: 12/28/2024] Open
Abstract
High sugar, high-fat diets and unhealthy lifestyles have led to an epidemic of obesity and obesity-related metabolic diseases, seriously placing a huge burden on socio-economic development. A deeper understanding and elucidation of the specific molecular biological mechanisms underlying the onset and development of obesity has become a key to the treatment of metabolic diseases. Recent studies have shown that the changes of bile acid composition are closely linked to the development of metabolic diseases. Bile acids can not only emulsify lipids in the intestine and promote lipid absorption, but also act as signaling molecules that play an indispensable role in regulating bile acid homeostasis, energy expenditure, glucose and lipid metabolism, immunity. Disorders of bile acid metabolism are therefore important risk factors for metabolic diseases. The farnesol X receptor, a member of the nuclear receptor family, is abundantly expressed in liver and intestinal tissues. Bile acids act as endogenous ligands for the farnesol X receptor, and erroneous FXR signaling triggered by bile acid dysregulation contributes to metabolic diseases, including obesity, non-alcoholic fatty liver disease and diabetes. Activation of FXR signaling can reduce lipogenesis and inhibit gluconeogenesis to alleviate metabolic diseases. It has been found that intestinal FXR can regulate hepatic FXR in an organ-wide manner. The crosstalk between intestinal FXR and hepatic FXR provides a new idea for the treatment of metabolic diseases. This review focuses on the relationship between bile acids and metabolic diseases and the current research progress to provide a theoretical basis for further research and clinical applications.
Collapse
Affiliation(s)
| | | | | | | | - Yuxia Xiong
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
46
|
Zheng W, Xu L, Jin M, Wang J, Rietjens IMCM. Effects of lambda-cyhalothrin on gut microbiota and related bile acid metabolism in mice. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136417. [PMID: 39536348 DOI: 10.1016/j.jhazmat.2024.136417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Since the gut microbiota plays a crucial role in host metabolism and homeostasis, its alterations induced by xenobiotics such as pesticides, could pose a risk to host health. The pyrethroid insecticides were frequently detected in surface water (up to 13 mg/L worldwide), sediments, and agricultural products; additionally, some previous studies indicated that pyrethroid insecticides could cause disruption of gut homeostasis. Hence herein, the normally used pyrethroid lambda-cyhalothrin (LCT) was selected and studied for its effects on the intestinal microbial community and its related bile acid metabolism using mice as the model species. Results showed that the total amount of bile acids in plasma and fecal samples from LCT treated mice markedly increased compared to controls, which could be mainly ascribed to the significantly raised proportions of taurine conjugated bile acids in plasma, and the increase in fecal secondary bile acids. In gut microbial profiles, a significantly increased richness of Prevotellacea and a depletion of Lachnospiraceae were found at the family level upon the treatment with lambda-cyhalothrin. In conclusion, results obtained on bacterial and bile acid profiles corroborate that the treatment of mice with LCT could affect gut microbial community with accompanying changes in bile acid homeostasis.
Collapse
Affiliation(s)
- Weijia Zheng
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands; Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Lingyuan Xu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Maojun Jin
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Jing Wang
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China; Sanya National Nanfan Research Institute of the Chinese Academy of Agricultural Sciences, Sanya 572024, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China.
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| |
Collapse
|
47
|
Kumar D, Poša M. Thermodynamics of Micelle Formation of Selected Homologous 7-Alkyl Derivatives of Na-Cholate in Aqueous Solution: Steroid Skeleton and the Alkyl Chain Conformation. Int J Mol Sci 2024; 25:13055. [PMID: 39684766 DOI: 10.3390/ijms252313055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Bile acid salts are steroid biosurfactants that build relatively small micelles compared to surfactants with an alkyl chain due to the rigid conformation of the steroid skeleton. In order to increase the capacity of micellar solubilization of the hydrophobic molecular guest, certain C7 alkyl derivatives were synthesized. Namely, introducing an alkyl group in the C7 position of the steroid skeleton results in a more effective increase in the micelle's hydrophobic domain (core) than the introduction in the C3 position. In comparison, fewer synthetic steps are required than if alkyl groups are introduced into the C12 position of cholic acid in the Grignard reaction. Here, the thermodynamic parameters of micellization (demicellization) of C7 alkyl (number of C atoms in the alkyl group: 2, 3, 4, and 8) derivatives of cholic acid anion in an aqueous solution without additives are examined (which have not yet been determined) in the temperature interval T (10-40) °C. The critical micellar concentration and the change in the standard molar enthalpy of demicellization (∆hdemic0) are determined by isothermal calorimetric titration (ICT). From the temperature dependence of ∆hdemic0, the change in the standard molar heat capacity of demicellization is obtained (∆Cdemic0), the value of which is proportional to the hydrophobic surface of the monomer, which in the micellar state is protected from hydrophobic hydration. The values of ∆Cdemic0 indicate that in the case of C7-alkyl derivatives of cholic acid anion with butyl and octyl chains, parts of the steroid skeleton and alkyl chain remain shielded from hydration after disintegration of the micelle. Conformational analysis can show that starting from the C7 butyl chain in the alkyl chain, sequences with gauche conformation are also possible without the formation of steric repulsive strain between the alkyl chain and the steroid skeleton so that the C7 alkyl chain takes an orientation above the convex surface of the steroid skeleton instead of an elongated conformation toward the aqueous solution. This is a significant observation, namely, if the micelle is used as a carrier of a hydrophobic drug and after the breakdown of the micelle in the biological system, the released drug has a lower tendency to associate with the monomer if its hydrophobic surface is smaller, i.e., the alkyl chain is oriented towards the angular methyl groups of the steroid skeleton (the ideal monomer increases the hydrophobic domain of the micelle, but in aqueous solution, it adopts a conformation with the as small hydrophobic surface as possible oriented towards the aqueous solution)-which then does not disturb the passage of the drug through the cell membrane.
Collapse
Affiliation(s)
- Dileep Kumar
- Laboratory for Chemical Computation and Modeling, Institute for Computational Science and Artificial Intelligence, Van Lang University, Ho Chi Minh City 70000, Vietnam
- Faculty of Applied Technology, School of Technology, Van Lang University, Ho Chi Minh City 70000, Vietnam
| | - Mihalj Poša
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, Hajduk Veljka 3, 21000 Novi Sad, Serbia
| |
Collapse
|
48
|
Antony F, Brough Z, Orangi M, Al-Seragi M, Aoki H, Babu M, Duong van Hoa F. Sensitive Profiling of Mouse Liver Membrane Proteome Dysregulation Following a High-Fat and Alcohol Diet Treatment. Proteomics 2024; 24:e202300599. [PMID: 39313981 DOI: 10.1002/pmic.202300599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024]
Abstract
Alcohol consumption and high-fat (HF) diets often coincide in Western society, resulting in synergistic negative effects on liver function. Although studies have analyzed the global protein expression in the context of alcoholic liver disease (ALD) and metabolic dysfunction-associated steatotic liver disease (MASLD), none has offered specific insights on liver dysregulation at the membrane proteome level. Membrane-specific profiling of metabolic and compensatory phenomena is usually overshadowed in conventional proteomic workflows. In this study, we use the Peptidisc method to isolate and compare the membrane protein (MP) content of the liver with its unique biological functions. From mice fed with an HF diet and ethanol in drinking water, we annotate over 1500 liver proteins with half predicted to have at least one transmembrane segment. Among them, we identify 106 integral MPs that are dysregulated compared to the untreated sample. Gene Ontology analysis reveals several dysregulated membrane-associated processes like lipid metabolism, cell adhesion, xenobiotic processing, and mitochondrial membrane formation. Pathways related to cholesterol and bile acid transport are also mutually affected, suggesting an adaptive mechanism to counter the upcoming steatosis of the liver model. Taken together, our Peptidisc-based profiling of the diet-dysregulated liver provides specific insights and hypotheses into the role of the transmembrane proteome in disease development, and flags desirable MPs for therapeutic and diagnostic targeting.
Collapse
Affiliation(s)
- Frank Antony
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zora Brough
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mona Orangi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mohammed Al-Seragi
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hiroyuki Aoki
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Mohan Babu
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Franck Duong van Hoa
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
49
|
Feng Y, Liu CH, Yang J, Zhang H, Li L, Yang Q, Gan W, Yang Z, Gong P, Fu C, Qian G, Li D. Integrative analysis of non12-hydroxylated bile acid revealed the suppressed molecular map of alternative pathway in nonalcoholic steatohepatitis mice. FASEB J 2024; 38:e70167. [PMID: 39556333 DOI: 10.1096/fj.202401630r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024]
Abstract
Bile acids (BAs) are significantly altered in the liver and serum of patients with nonalcoholic steatohepatitis (NASH). However, the underlying mechanisms of these changes, particularly BA alternative pathways (BAP) responsible for non12-OH BAs, remain unclear. RNA-seq data were initially analyzed to reveal the changes of gene expression in NASH patients. Targeted metabolomics were conducted on plasma from NASH mice induced by high-fat or western diet with CCl4 for 10-24 weeks. Liver tissues were examined using proteomics, RT-qPCR, and western blotting. An integrated approach was then employed to analyze protein interactions and network correlations. Analysis of RNA-seq data revealed the inhibition of CYP7B1 in NASH patients, indicating the dysregulation of BAP. In NASH mouse models, dysregulation of BA circulation was observed by increased plasma total BA (TBA) levels and decreased liver TBA, with liver swelling and histopathological changes. Targeted metabolomics revealed suppressed levels of non12-OH BAs, which inversely correlated with increased liver injury markers. The reduced mRNA and protein expression of Fxr and upregulation of Lxr signaling in livers suggested the suppressed BAP was modulated by Fxr-Lxr signaling. Moreover, BAP interactions predominantly implicated multiple metabolism disruptions, involving 7 hub proteins (Hk1, Acadsb, Pklr, Insr, Ldlr, Cyp27a1, and Cyp7b1), offering promising therapeutic targets for NASH. We presented the metabolic and proteomic map of BAP and its regulatory network in NASH progression. Therapeutic targeting of BAP or its co-regulatory proteins holds promise for NASH treatment and metabolic syndrome management.
Collapse
Affiliation(s)
- Yanruyu Feng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
- Ninth People's Hospital of Zhengzhou, Zhengzhou, China
| | - Chang-Hai Liu
- Center of Infectious Diseases, Division of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jingtao Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - He Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Qian Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Wei Gan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zi Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Puyang Gong
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Chunmei Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Guangsheng Qian
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Dapeng Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
An P, Fan Y, Wang Q, Huang N, Chen H, Sun J, Du Z, Zhang C, Li J. Cholic acid activation of GPBAR1 does not induce or exacerbate acute pancreatitis but promotes exocrine pancreatic secretion. Biochem Biophys Res Commun 2024; 735:150825. [PMID: 39426134 DOI: 10.1016/j.bbrc.2024.150825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/22/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Obstruction of bile ducts due to gallstones can lead to biliary acute pancreatitis (BAP). According to Perides et al., G protein-coupled bile acid receptor-1 (GPBAR1) mediates BAP. However, Zi's findings suggest that GPR39, rather than GPBAR1, mediates TLCAS-induced increases in cytosolic calcium and acinar cell necrosis, casting doubt on the role of GPBAR1 in BAP. Numerous G protein-coupled receptors on pancreatic acinar cells utilize Ca2+ and cyclic adenosine monophosphate (cAMP) as second messengers to manage pancreatic exocrine secretion, with significant cross-talk between these signals. The primary bile acid cholic acid (CA) and its conjugated forms are predominant in the human gallbladder. This study aimed to clarify the role and physiological significance of GPBAR1 by investigating the physiological and pathological effects of CA activation on GPBAR1 in pancreatic acinar cells. Isolated rat pancreatic acinar cells were treated with CA and CCK in vitro to observe the effect of CA-induced cAMP signaling on CCK-induced physiological and pathological calcium signaling. In vivo evaluations involved reverse biliopancreatic duct injections of 5 % sodium taurocholate (STC) or 5 % CA in rats. CA induced intracellular cAMP signaling in a concentration-dependent manner without increasing the intracellular Ca2+ concentration. CA did not independently cause calcium overload or enzyme activation, nor did it exacerbate calcium overload or enzyme activation from high-dose CCK. Reverse biliopancreatic duct injections of 5 % CA did not cause acute pancreatitis in the rats. Transcriptomic analysis revealed that 50 μM CA induced changes in gene expression related to protein synthesis in the endoplasmic reticulum and ribosomes. Furthermore, 50 μM CA accelerated the calcium waves and increased the enzyme secretion induced by CCK. GPBAR1 was found on the basolateral membrane in rat pancreatic tissue rather than near the apical region of acinar cells. GPBAR1 activation is not crucial for BAP activity but may play a role in bile acid regulation of pancreatic exocrine secretion, suggesting that GPBAR1 is a potential therapeutic target for pancreatic exocrine insufficiency.
Collapse
Affiliation(s)
- Peng An
- Department of Integrated Chinese Traditional and Western Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| | - Yudan Fan
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| | - Qian Wang
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| | - Na Huang
- National & Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| | - Haiyan Chen
- Core Research Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| | - Jin Sun
- National & Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| | - Ziwei Du
- National & Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| | - Chen Zhang
- National & Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| | - Jun Li
- National & Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| |
Collapse
|