1
|
Yu L, Shi X, Lu X, Zhao X, Yu Y, Fang Y, Wang D. Rehmannia glutinosa polysaccharides alleviate cyclophosphamide-induced immunosuppression by enhancing immunity and restoring intestinal homeostasis. Int J Biol Macromol 2025; 311:143692. [PMID: 40316077 DOI: 10.1016/j.ijbiomac.2025.143692] [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: 03/02/2025] [Revised: 04/16/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
This study investigated the immunomodulatory effects of Rehmannia glutinosa polysaccharides (RGP) in a cyclophosphamide (CTX)-induced immunosuppression model. RGP treatment alleviated weight loss, immune organ atrophy, and hematological abnormalities, restored B and T cell populations, and enhanced the immune function. It strengthened intestinal barrier integrity by upregulating tight junction proteins, increasing goblet cell numbers, and promoting IgA secretion, while also reducing pro-inflammatory cytokines and increasing IL-10 levels. RGP further regulated the gut microbiota composition, increasing beneficial bacteria and reducing pathogens, while promoting SCFA production and improving metabolic homeostasis. These findings highlight RGP as a potential natural immunomodulator, supporting further research into its molecular mechanisms and clinical applications.
Collapse
Affiliation(s)
- Lin Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xiaofeng Shi
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xuanqi Lu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xuping Zhao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yaming Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yushan Fang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
2
|
Luo X, Ma R, Cai H, Su X, Wang S, Qiao T. Formation of non-cholesterol gallstones in populations within clonorchiasis endemic regions is closely related to Clonorchis sinensis infection. BMC Gastroenterol 2025; 25:345. [PMID: 40340656 PMCID: PMC12060310 DOI: 10.1186/s12876-025-03939-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/25/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Numerous risk factors are linked to gallbladder stone disease (GBSD). Nonetheless, the relationship between Clonorchis sinensis (C. sinensis) and this condition remains to be clarified. METHODS The antibody against C. sinensis in serum and the glucose, triglyceride, and cholesterol levels were investigated in 220 patients with GBSD and 251 controls. Bile components were analysed in patients with gallbladder polyps (GP, n = 18), gallstones (GS, n = 265), and GS combined with C. sinensis infection (GSI, n = 243). Additionally, the gallbladder ejection fraction (%E), residual gallbladder volume (RGV) at 1 h after a fatty meal, and fasting gallbladder volume (FGV) were compared among the GP (n = 43), GS (n = 311), and GSI (n = 277) groups. RESULTS The results indicated positive antibody against C. sinensis (OR: 1.759, 95% CI: 1.163-2.662) and hyperglycaemia (glucose concentration ≥ 6.10 mmol/L, OR: 2.263, 95% CI: 1.227-4.172) were risk factors for GBSD. There were more non-cholesterol stones in GSI patients (216/241, 89.6%) than in GS patients (137/281, 48.8%) (P < 0.0001). Microscopic observations revealed that mucus containing glycogen coated the C. sinensis eggs and the proportion of dead eggs gradually increased in bile, sediment, and stones alongside rising calcium salt content. Total bile acid and cholesterol concentrations were lower in GSI patients than in GP patients or GS patients (P < 0.05). Moreover, increased FGV and RGV and decreased %E were observed in GSI patients compared with GP patients (P < 0.001). CONCLUSIONS The formation of non-cholesterol gallstones in populations residing in endemic areas is related to the deposition, death, and calcification of eggs in the gallbladder, changes in bile components, and decreased gallbladder motility caused by C. sinensis infection.
Collapse
Affiliation(s)
- Xiaobing Luo
- Department of Clinical Laboratory, Guangzhou Nansha People's Hospital, No. 7 Xingye Road, Dagang Town, Nansha District, Guangzhou, Guangdong, China.
| | - Ruihong Ma
- Department of Clinical Laboratory, Guangzhou Nansha People's Hospital, No. 7 Xingye Road, Dagang Town, Nansha District, Guangzhou, Guangdong, China
| | - Hongying Cai
- Department of Pathology, Guangzhou Nansha People's Hospital, No. 7 Xingye Road, Dagang Town, Nansha District, Guangzhou, Guangdong, China
| | - Xiangyu Su
- Department of Clinical Laboratory, Guangzhou Nansha People's Hospital, No. 7 Xingye Road, Dagang Town, Nansha District, Guangzhou, Guangdong, China
| | - Sangui Wang
- Dongguan Nancheng Hospital, No. 55 Nancheng Avenue, Guantai Road, Dongguan, Guangdong, China.
| | - Tie Qiao
- The Second People's Hospital of Guangzhou Panyu District, No. 88 Gangdong Road, Dashi Street, Panyu District, Guangzhou, Guangdong, China.
| |
Collapse
|
3
|
Xu Z, Pei Y, Wang H, Li X. Comparative analysis of gut microbiota-mediated bile acid profiles in Bufo gargarizans and Rana chensinensis tadpoles. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101530. [PMID: 40373385 DOI: 10.1016/j.cbd.2025.101530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/17/2025]
Abstract
Bile acids (BAs) are cholesterol derivatives synthesized by the liver, exhibit variation between different species. Researchers have long appreciated that microbiota play the roles in the biotransformation of BAs. However, relatively few studies have been reported on microbial-mediated production and transformation of BAs in amphibians. Our focus here is principally on difference of intestinal microbial diversity and BAs profiles between two common amphibians, Bufo gargarizans (B. gargarizans) and Rana chensinensis (R. chensinensis) tadpoles, through intestinal targeted BAs metabolomics and fecal metagenomic sequencing. The results demonstrated that B. gargarizans possessed higher levels of total BAs and higher ratio of unconjugated / conjugated BAs. In addition, the relative abundance of microbiota with bile salt hydrolase (BSH) activity in B. gargarizans was significantly higher than that of R. chensinensis, which may facilitate the conversion of conjugated to unconjugated BAs. Meanwhile the higher prevalence of bile-acid-induced (BAI) gene encoding microbiota in R. chensinensis may promote the synthesis of deoxycholic acid (DCA). Furthermore, discrepancies in virulence factors (VFs) and energy metabolism were observed between the two species, which may be linked to differences in the microbiota. This study revealed substantial differences in intestinal microbes and BAs across amphibian species, emphasizing the significant impact of intestinal microbes on BAs metabolism.
Collapse
Affiliation(s)
- Zhangying Xu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yuebin Pei
- Cotton Research Institute, Shanxi Agriculture University, Yuncheng, Shanxi 044000, China
| | - Hongyuan Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xinyi Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
4
|
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
|
5
|
Tessier MEM, Shneider BL, Petrosino JF, Preidis GA. Bile acid and microbiome interactions in the developing child. J Pediatr Gastroenterol Nutr 2025; 80:832-839. [PMID: 39959949 PMCID: PMC12068970 DOI: 10.1002/jpn3.70014] [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: 04/26/2024] [Revised: 12/06/2024] [Accepted: 12/23/2024] [Indexed: 05/13/2025]
Abstract
Interactions between the gut microbiome and bile acids are complex and are linked to outcomes in pediatric liver disease by mechanisms that are incompletely understood. In adults, primary bile acids are synthesized in the liver and secreted into the intestine, where complex communities of gut microbes deconjugate, oxidize, epimerize, and 7α-dehydroxylate bile acids into a diverse array of unconjugated, secondary, allo-, iso-, and oxo-bile acids. In contrast, the infant gut microbiota contains a simple, Bifidobacterium-dominant community that transitions to a more diverse, adult-like community as additional microbes colonize the gut. This microbial succession gradually confers deconjugation, oxidation, epimerization, and 7α-dehydroxylation activities that mature the bile acid pool from a profile dominated by primary bile acids early in life to a more diverse, adult-like bile acid profile in later childhood. Altered bile acid profiles in pediatric cholestatic disorders have the potential to change the developmental trajectory of the microbiome. Conversely, alterations in the gut microbiome may re-shape the bile acid pool and hepatic bile acid metabolism. Understanding the mechanisms underlying these interactions will increase our understanding of liver pathophysiology and will motivate new therapeutic strategies for pediatric hepatic disorders. This review aims to highlight differences between the pediatric and adult intestinal microbiome and bile acid pool, and to discuss interactions between gut microbes and bile acids that are critical in early life and that may impact outcomes in infants and children with cholestatic liver disease, including biliary atresia.
Collapse
Affiliation(s)
- Mary Elizabeth M. Tessier
- Department of Pediatrics, Section of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine/ Texas Children’s Hospital, Houston, TX, United States
| | - Benjamin L. Shneider
- Department of Pediatrics, Section of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine/ Texas Children’s Hospital, Houston, TX, United States
| | - Joseph F. Petrosino
- Department of Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Geoffrey A Preidis
- Department of Pediatrics, Section of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine/ Texas Children’s Hospital, Houston, TX, United States
| |
Collapse
|
6
|
Qi Z, Zhang W, Zhang P, Qu Y, Zhong H, Zhou L, Zhou W, Yang W, Xu H, Zhao X, Wu H, Qian J, Ge J. The gut microbiota-bile acid-TGR5 axis orchestrates platelet activation and atherothrombosis. NATURE CARDIOVASCULAR RESEARCH 2025; 4:584-601. [PMID: 40217125 DOI: 10.1038/s44161-025-00637-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 03/12/2025] [Indexed: 05/18/2025]
Abstract
Gut microbiota-derived bile acids are crucial in the pathogenesis and treatment of metabolic diseases. However, their impact on platelet activation and thrombosis in coronary artery disease (CAD) remains poorly understood. In this study, we observed reduced serum deoxycholic acid (DCA) in patients with CAD and an underrepresentation of Bacteroides vulgatus in the gut microbiota of patients with CAD, affecting DCA metabolism. We used Takeda G-protein-coupled receptor 5 (TGR5) inhibitors and TGR5 knockout mice to show that DCA inhibited agonist-induced platelet activation and thrombosis by interacting with the platelet TGR5. Oral gavage treatments with DCA, B. vulgatus and stool from healthy individuals suppressed platelet hyperreactivity and thrombosis in atherosclerotic ApoE-/- mice, reduced microvascular thrombosis and protected the heart from myocardial ischemia/reperfusion injury. Here we describe the role of the bile acid DCA in platelet activation and suggest that targeting the gut microbiota and/or altering bile acid metabolism may be beneficial to treat CAD-associated thrombosis.
Collapse
Affiliation(s)
- Zhiyong Qi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Wei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Peng Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Yanan Qu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Haoxuan Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Luning Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Wenxuan Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Wenlong Yang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Huajie Xu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xin Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Hongyi Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China.
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Ghaffari MH, Sauerwein H, Sadri H, Schuchardt S, Martín-Tereso J, Doelman JH, Daniel JB. Longitudinal characterization of the metabolome of dairy cows transitioning from one lactation to the next: Investigations in fecal samples. J Dairy Sci 2025; 108:5405-5419. [PMID: 40043758 DOI: 10.3168/jds.2025-26273] [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: 01/06/2025] [Accepted: 01/31/2025] [Indexed: 05/03/2025]
Abstract
The fecal metabolome comprises metabolites that are excreted or not absorbed by the animal. This study examined the changes in the fecal metabolome of dairy cows from the end of one lactation period, through the dry period, and into the subsequent lactation. Twelve Holstein cows (BW = 745 ± 71 kg, BCS = 3.43 ± 0.66) were housed in a tiestall barn from 7 wk before to 15 wk after parturition, with dry-off occurring approximately 6 wk before the expected calving date (mean dry-off time = 42 d). Fecal samples were taken at wk -7, -5, -1, +1, +5, +10, and +15 relative to calving. Targeted metabolomics identified a total of 93 metabolites, including AA, biogenic amines, bile acids (BA), acylcarnitines (AcylCN), and some phospholipids. Principal component analysis (PCA) revealed clear metabolic shifts that showed a clear separation between the samples from the dry period and the samples from the end, early, and middle of lactation, indicating significant changes in the metabolic profiles in the feces. The transition from the dry period (wk -5, -1 relative to calving) to lactation (wk +1, +5, +10, +15, and -7 relative to calving) is characterized by an increase in fecal AA and metabolites, such as Glu, Met, β-alanine, and methionine sulfoxide, reflecting a shift in nitrogen metabolism to support increased protein metabolism for milk production. Higher concentrations of polyamines, such as spermidine and putrescine, were observed postpartum, indicating increased cell growth and improved tissue regeneration. Elevated gamma-aminobutyric acid levels during lactation indicate increased microbial activity driven by a nutrient-rich diet. Results showed significant adjustments in BA profiles as cows transitioned into lactation. Deoxycholic acid remained the predominant BA in feces, reflecting ongoing microbial transformation, whereas glycine- and taurine-conjugated BA increased postpartum, suggesting improved enterohepatic circulation and lipid absorption. Fecal AcylCN showed dynamic shifts with elevated levels during late gestation, a decrease in the dry period, and an increase postpartum, indicating increased fatty acid oxidation to meet energy demands. Results showed that phosphatidylcholines decreased prepartum but increased after calving. This indicates shifts in lipid metabolism reflecting energy requirements in lactation and suggests that fecal lipid composition is an indicator of metabolic adaptations in dairy cows. In particular, PCA revealed considerable overlap in the fecal metabolite profiles of multiparous and primiparous cows, indicating similar metabolic profiles. This was also confirmed by volcano plots, which showed no significant differences in fecal metabolism between the 2 groups across different weeks relative to calving (wk -7, -5, -1, +1, +5, +10, and +15). Overall, these results emphasize the complex interactions between dietary factors, liver and gastrointestinal function, and the gut microbiome in shaping the fecal metabolite profile of dairy cows. These results underscore the value of this data set in advancing the application of fecal metabolome profiling to investigate metabolic changes during critical transitions in the lactation cycle of dairy cows.
Collapse
Affiliation(s)
- M H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| | - H Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - H Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, 5166616471 Tabriz, Iran
| | - S Schuchardt
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | | | - J H Doelman
- Trouw Nutrition R&D, 3800 AG, Amersfoort, the Netherlands
| | - J B Daniel
- Trouw Nutrition R&D, 3800 AG, Amersfoort, the Netherlands.
| |
Collapse
|
8
|
He Q, Li X, Li H, Tan A, Chi Y, Fang D, Li X, Liu Z, Shang Q, Zhu Y, Cielecka-Piontek J, Chen J. TGR5 Activation by Dietary Bioactives and Related Improvement in Mitochondrial Function for Alleviating Diabetes and Associated Complications. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6293-6314. [PMID: 40045496 DOI: 10.1021/acs.jafc.4c10395] [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: 03/20/2025]
Abstract
Takeda G protein-coupled receptor 5 (TGR5), also known as G protein-coupled bile acid receptor 1 (GPBAR1), is a cell surface receptor involved in key physiological processes, including glucose homeostasis and energy metabolism. Recent research has focused on the role of TGR5 activation in preventing or treating diabetes while also highlighting its potential impact on the progression of diabetic complications. Functional foods and edible plants have emerged as valuable sources of natural compounds that can activate TGR5, offering potential therapeutic benefits for diabetes management. Despite growing interest, studies on the activation of TGR5 by dietary bioactive compounds remain scattered. This Review aims to provide a comprehensive analysis of how dietary bioactives act as potential agents for TGR5 activation in managing diabetes and its complications. It explores the mechanisms of TGR5 activation through both direct agonistic effects and indirect pathways via modulation of the gut microbiota and bile acid metabolism.
Collapse
Affiliation(s)
- Quanrun He
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Xinhang Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Haimeng Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Aditya Tan
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Yunlin Chi
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
| | - Daozheng Fang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Xinyue Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Zhihao Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Qixiang Shang
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Yong Zhu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznan, Poland
| | - Jihang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Boulevard, Longgang District, Shenzhen, Guangdong 518172, P.R. China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, Shenzhen-Hong Kong International Science and Technology Park, No. 3 Binglang Road, Futian Free Trade Zone, Futian District, Shenzhen, Guangdong 518045, P.R. China
| |
Collapse
|
9
|
Xie Y, Chen Q, Shan D, Pan X, Hu Y. Unraveling the role of the gut microbiome in pregnancy disorders: insights and implications. Front Cell Infect Microbiol 2025; 15:1521754. [PMID: 40125520 PMCID: PMC11925892 DOI: 10.3389/fcimb.2025.1521754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
The gut microbiota is the collective term for the microorganisms that reside in the human gut. In recent years, advances in sequencing technology and bioinformatics gradually revealed the role of gut microbiota in human health. Dramatic changes in the gut microbiota occur during pregnancy due to hormonal and dietary changes, and these changes have been associated with certain gestational diseases such as preeclampsia (PE) and gestational diabetes mellitus (GDM). Modulation of gut microbiota has also been proposed as a potential treatment for these gestational diseases. The present article aims to review current reports on the association between gut microbiota and gestational diseases, explore possible mechanisms, and discuss the potential of probiotics in gestational diseases. Uncovering the link between gut microbiota and gestational diseases could lead to a new therapeutic approach.
Collapse
Affiliation(s)
- Yupei Xie
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Qian Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Dan Shan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Xiongfei Pan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- West China Second University Hospital, Sichuan University, Shuangliu Institute of Women’s and Children’s Health, Shuangliu Maternal and Child Health Hospital, Chengdu, China
| | - Yayi Hu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| |
Collapse
|
10
|
Zhang J, Shen M, Yin Y, Chen Y, Deng X, Mo J, Zhou X, Lin J, Chen X, Xie X, Wu X, Chen X. Carnosic acid reduces lipid content, enhances gut health, and modulates microbiota composition and metabolism in diet-induced obese mice. Food Funct 2025; 16:1888-1902. [PMID: 39932492 DOI: 10.1039/d4fo04534c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Carnosic acid (CA) is a bioactive phenolic diterperne compound found in sage and rosemary. The present study investigated the beneficial effects of CA (50 and 100 mg per kg bw) in diet-induced obese mice and the underlying mechanisms of action. After the intervention, the physiology, lipid metabolism, and tissue morphology, as well as the inflammation, gut microbiota, and metabolomics in the colon were measured. We found that CA improved the composition and metabolism of the gut microbiota in obese mice, with Akkermansia being the dominant bacterium negatively correlated with obesity and various fecal metabolites. Regarding the intestinal barrier function, CA promoted the expression of tight junction proteins and inhibited the TLR4/MyD88/NF-κB signaling pathway in obese mice to alleviate colonic inflammation. These results suggest that CA improved multiple aspects of gut health in diet-induced obesity in mice, providing a scientific basis for future clinical studies in humans.
Collapse
Affiliation(s)
- Jing Zhang
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Mengzhu Shen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yue Yin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yuru Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xianying Deng
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Jingyun Mo
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xiaoling Zhou
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Juanying Lin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinxin Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinwei Xie
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xian Wu
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, Ohio 45056, USA.
| | - Xuexiang Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| |
Collapse
|
11
|
Dias BDC, Lamarca AP, Machado DT, Kloh VP, de Carvalho FM, Vasconcelos ATR. Metabolic pathways associated with Firmicutes prevalence in the gut of multiple livestock animals and humans. Anim Microbiome 2025; 7:20. [PMID: 40033444 PMCID: PMC11874851 DOI: 10.1186/s42523-025-00379-y] [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: 07/31/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
Dynamic interspecific interactions and environmental factors deeply impact the composition of microbiotic communities in the gut. These factors intertwined with the host's genetic background and social habits cooperate synergistically as a hidden force modulating the host's physiological and health determinants, with certain bacterial species being maintained from generation to generation. Firmicutes, one of the dominant bacterial phyla present across vertebrate classes, exhibits a wide range of functional capabilities and colonization strategies. While ecological scenarios involving microbial specialization and metabolic functions have been hypothesized, the specific mechanisms that sustain the persistence of its microbial taxa in a high diversity of hosts remain elusive. This study fills this gap by investigating the Firmicutes metabolic mechanisms contributing to their prevalence and heritability in the host gut on metagenomes-assembled bacterial genomes collected from 351 vertebrate samples, covering 18 food-producing animals and humans, specific breeds and closely-related species. We observed that taxa belonging to Acetivibrionaceae, Clostridiaceae, Lachnospiraceae, Ruminococcaceae, and the not well understood CAG-74 family were evolutionarily shared across all hosts. These prevalent taxa exhibit metabolic pathways significantly correlated with extra-host survival mechanisms, cell adhesion, colonization and host transmission, highlighted by sporulation, glycan biosynthesis, bile acid metabolism, and short-chain fatty acid encoded genes. Our findings provide a deeper understanding of the ecological foundations governing distinct transmission modes, effective colonization establishment, and maintenance of Firmicutes, offering new perspectives on both well-known and poorly characterized species.
Collapse
Affiliation(s)
- Beatriz do Carmo Dias
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Brazil
| | - Alessandra Pavan Lamarca
- Laboratório de Bioinformática e Evolução Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas Terra Machado
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Brazil
| | - Vinicius Prata Kloh
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Brazil
| | | | | |
Collapse
|
12
|
Zhu TF, Guo HP, Nie L, Chen J. Oral administration of LEAP2 enhances immunity against Edwardsiella tarda through regulation of gut bacterial community and metabolite in mudskipper. FISH & SHELLFISH IMMUNOLOGY 2025; 158:110128. [PMID: 39824300 DOI: 10.1016/j.fsi.2025.110128] [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: 10/22/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
The liver-expressed antimicrobial peptide 2 (LEAP2) is gaining recognition for its immune regulatory functions beyond direct antimicrobial activity. In this study, we investigated the role of mudskipper (Boleophthalmus pectinirostris) LEAP2 (BpLEAP2) in enhancing the survival, gut health, and immune resilience against Edwardsiella tarda infection. Pre-oral delivery of BpLEAP2 significantly improved survival rates and mitigated infection-induced damage to the gut, as evidenced by preserved villus length and goblet cell count. Analysis of gut microbial communities using 16S rRNA sequencing revealed that pre-oral delivery of BpLEAP2 increased microbial diversity, evenness, and the abundance of beneficial genera such as Pseudoalteromonas and Shewanella, while reducing pathogenic genera like Pseudorhodobacter. Metabolomic profiling showed that BpLEAP2 altered the gut metabolite composition, significantly increasing levels of bile acids and amino acids, which are known to support gut health and immune responses. Correlation analysis demonstrated strong positive associations between BpLEAP2-induced microbial shifts and increased metabolites involved in amino acid metabolism. These findings suggest that BpLEAP2 promotes intestinal homeostasis by modulating gut microbiota composition and enhancing beneficial metabolite production, ultimately improving gut barrier integrity and conferring resistance against E. tarda infection. This study highlights the potential application of BpLEAP2 in enhancing disease resilience in aquaculture species, offering a promising strategy for sustainable aquaculture practices.
Collapse
Affiliation(s)
- Ting-Fang Zhu
- State Key Laboratory for Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Hai-Peng Guo
- State Key Laboratory for Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China
| | - Li Nie
- State Key Laboratory for Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| | - Jiong Chen
- State Key Laboratory for Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
13
|
Ekwudo MN, Gubert C, Hannan AJ. The microbiota-gut-brain axis in Huntington's disease: pathogenic mechanisms and therapeutic targets. FEBS J 2025; 292:1282-1315. [PMID: 38426291 PMCID: PMC11927060 DOI: 10.1111/febs.17102] [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: 10/13/2023] [Revised: 01/08/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
Huntington's disease (HD) is a currently incurable neurogenerative disorder and is typically characterized by progressive movement disorder (including chorea), cognitive deficits (culminating in dementia), psychiatric abnormalities (the most common of which is depression), and peripheral symptoms (including gastrointestinal dysfunction). There are currently no approved disease-modifying therapies available for HD, with death usually occurring approximately 10-25 years after onset, but some therapies hold promising potential. HD subjects are often burdened by chronic diarrhea, constipation, esophageal and gastric inflammation, and a susceptibility to diabetes. Our understanding of the microbiota-gut-brain axis in HD is in its infancy and growing evidence from preclinical and clinical studies suggests a role of gut microbial population imbalance (gut dysbiosis) in HD pathophysiology. The gut and the brain can communicate through the enteric nervous system, immune system, vagus nerve, and microbiota-derived-metabolites including short-chain fatty acids, bile acids, and branched-chain amino acids. This review summarizes supporting evidence demonstrating the alterations in bacterial and fungal composition that may be associated with HD. We focus on mechanisms through which gut dysbiosis may compromise brain and gut health, thus triggering neuroinflammatory responses, and further highlight outcomes of attempts to modulate the gut microbiota as promising therapeutic strategies for HD. Ultimately, we discuss the dearth of data and the need for more longitudinal and translational studies in this nascent field. We suggest future directions to improve our understanding of the association between gut microbes and the pathogenesis of HD, and other 'brain and body disorders'.
Collapse
Affiliation(s)
- Millicent N. Ekwudo
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| | - Anthony J. Hannan
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
- Department of Anatomy and PhysiologyUniversity of MelbourneParkvilleAustralia
| |
Collapse
|
14
|
Almheiri RT, Hajjar B, Alkhaaldi SMI, Rabeh N, Aljoudi S, Abd-Elrahman KS, Hamdan H. Beyond weight loss: exploring the neurological ramifications of altered gut microbiota post-bariatric surgery. J Transl Med 2025; 23:223. [PMID: 39994634 PMCID: PMC11852891 DOI: 10.1186/s12967-025-06201-2] [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: 12/03/2024] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
This review discusses findings related to neurological disorders, gut microbiota, and bariatric surgery, focusing on neurotransmitters, neuroendocrine, the pathophysiology of bacteria contributing to disorders, and possible therapeutic interventions. Research on neurotransmitters suggests that their levels are heavily influenced by gut microbiota, which may link them to neurological disorders such as Alzheimer's disease, Parkinson's disease, Multiple sclerosis, Depression, and Autism spectrum disorder. The pathophysiology of bacteria that reach and influence the central nervous system has been documented. Trends in microbiota are often observed in specific neurological disorders, with a prominence of pro-inflammatory bacteria and a reduction in anti-inflammatory types. Furthermore, bariatric surgery has been shown to alter microbiota profiles similar to those observed in neurological disorders. Therapeutic interventions, including fecal microbiota transplants and probiotics, have shown potential to alleviate neurological symptoms. We suggest a framework for future studies that integrates knowledge from diverse research areas, employs rigorous methodologies, and includes long-trial clinical control groups.
Collapse
Affiliation(s)
- Rashed T Almheiri
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Baraa Hajjar
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Saif M I Alkhaaldi
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Nadia Rabeh
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Sara Aljoudi
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates
| | - Khaled S Abd-Elrahman
- Department of Anesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
- Department of Medical Sciences, College of Medicine and Health Science, Khalifa University, 127788, Abu Dhabi, United Arab Emirates.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| | - Hamdan Hamdan
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, 127788, Abu Dhabi, United Arab Emirates.
- Healthcare Engineering Innovation Group (HEIG), Khalifa University of Science and Technology, 127788, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
15
|
Kumar S, Mukherjee R, Gaur P, Leal É, Lyu X, Ahmad S, Puri P, Chang CM, Raj VS, Pandey RP. Unveiling roles of beneficial gut bacteria and optimal diets for health. Front Microbiol 2025; 16:1527755. [PMID: 40041870 PMCID: PMC11877911 DOI: 10.3389/fmicb.2025.1527755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
The gut microbiome plays a pivotal role in human health, influencing digestion, immunity, and disease prevention. Beneficial gut bacteria such as Akkermansia muciniphila, Adlercreutzia equolifaciens, and Christensenella minuta contribute to metabolic regulation and immune support through bioactive metabolites like short-chain fatty acids (SCFAs). Dietary patterns rich in prebiotics, fermented foods, and plant-based bioactive compounds, including polyphenols and flavonoids, promote microbiome diversity and stability. However, challenges such as individual variability, bioavailability, dietary adherence, and the dynamic nature of the gut microbiota remain significant. This review synthesizes current insights into gut bacteria's role in health, emphasizing the mechanisms by which dietary interventions modulate microbiota. Additionally, it highlights advancements in microbiome-targeted therapies and the transformative potential of personalized nutrition, leveraging microbiota profiling and artificial intelligence (AI) to develop tailored dietary strategies for optimizing gut health and mitigating chronic inflammatory disorders. Addressing these challenges requires a multidisciplinary approach that integrates scientific innovation, ethical frameworks, and practical implementation strategies.
Collapse
Affiliation(s)
- Suresh Kumar
- National Institute of Biologicals, Ministry of Health & Family Welfare, Govt. of India, Noida, India
| | - Riya Mukherjee
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
| | - Pratibha Gaur
- Centre for Drug Design Discovery and Development (C4D), SRM University Delhi-NCR, Sonepat, India
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
| | - Élcio Leal
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| | - Xiaoming Lyu
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Ha'il, Saudi Arabia
| | - Paridhi Puri
- University Centre for Research and Development, Chandigarh University, Mohali, India
| | - Chung-Ming Chang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
- Master & Ph.D Program in Biotechnology Industry, Chang Gung University, Taoyuan, Taiwan
| | - V. Samuel Raj
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| | - Ramendra Pati Pandey
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| |
Collapse
|
16
|
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
|
17
|
Jiang SS, Kang ZR, Chen YX, Fang JY. The gut microbiome modulate response to immunotherapy in cancer. SCIENCE CHINA. LIFE SCIENCES 2025; 68:381-396. [PMID: 39235561 DOI: 10.1007/s11427-023-2634-7] [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: 04/14/2024] [Accepted: 06/05/2024] [Indexed: 09/06/2024]
Abstract
Gut microbiota have been reported to play an important role in the occurrence and development of malignant tumors. Currently, clinical studies have identified specific gut microbiota and its metabolites associated with efficacy of immunotherapy in multiple types of cancers. Preclinical investigations have elucidated that gut microbiota modulate the antitumor immunity and affect the efficacy of cancer immunotherapy. Certain microbiota and its metabolites may favorably remodel the tumor microenvironment by engaging innate and/or adaptive immune cells. Understanding how the gut microbiome interacts with cancer immunotherapy opens new avenues for improving treatment strategies. Fecal microbial transplants, probiotics, dietary interventions, and other strategies targeting the microbiota have shown promise in preclinical studies to enhance the immunotherapy. Ongoing clinical trials are evaluating these approaches. This review presents the recent advancements in understanding the dynamic interplay among the host immunity, the microbiome, and cancer immunotherapy, as well as strategies for modulating the microbiome, with a view to translating into clinical applications.
Collapse
Affiliation(s)
- Shan-Shan Jiang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Zi-Ran Kang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China.
| |
Collapse
|
18
|
Ionescu VA, Gheorghe G, Bacalbasa N, Diaconu CC. Metabolic Dysfunction-Associated Steatotic Liver Disease: Pathogenetic Links to Cardiovascular Risk. Biomolecules 2025; 15:163. [PMID: 40001466 PMCID: PMC11852489 DOI: 10.3390/biom15020163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is correlated with an increased cardiovascular risk, independent of other traditional risk factors. The mechanisms underlying this pathogenic link are complex yet remain incompletely elucidated. Among these, the most significant are visceral adiposity, low-grade inflammation and oxidative stress, endothelial dysfunction, prothrombotic status, insulin resistance, dyslipidemia and postprandial hyperlipemia, gut dysbiosis, and genetic mutations. Cardiovascular diseases are the leading cause of death in patients with MASLD. These patients have an increased incidence of coronary artery disease, carotid artery disease, structural and functional cardiac abnormalities, and valvulopathies, as well as arrhythmias and cardiac conduction disorders. In this review, we present the latest data on the association between MASLD and cardiovascular risk, focusing on the pathogenic mechanisms that explain the correlation between these two pathologies. Given the high rates of cardiovascular morbidity and mortality among patients with MASLD, we consider it imperative to raise awareness of the risks associated with this condition within the general population. Further research is essential to clarify the mechanisms underlying the increased cardiovascular risk linked to MASLD. This understanding may facilitate the identification of new diagnostic and prognostic biomarkers for these patients, as well as novel therapeutic targets.
Collapse
Affiliation(s)
- Vlad Alexandru Ionescu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania;
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Gina Gheorghe
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania;
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Nicolae Bacalbasa
- Department of Visceral Surgery, Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania;
- Department of Surgery, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania
| | - Camelia Cristina Diaconu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania;
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
- Academy of Romanian Scientists, 050085 Bucharest, Romania
| |
Collapse
|
19
|
Tang C, Pulliam C, Abiodun A, Parris A, Campbell A, Li J. Probing Antimicrobial Activity and Mechanism of Action of a Bile Acid-Derived Antibiotic. ACS OMEGA 2025; 10:1727-1734. [PMID: 39829505 PMCID: PMC11740247 DOI: 10.1021/acsomega.4c09804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
Antibiotics have revolutionized medicine, saving countless lives since the introduction of penicillin. However, antimicrobial resistance has challenged their efficacy, prompting ongoing efforts to develop new antibiotics. This study explores the antimicrobial effects of a bile acid derivative, BA-3/4-Butyl. By analyzing the interactions of BA-3/4-Butyl with model bacterial (DOPC/DOPG) and mammalian (DOPC/cholesterol) membranes and by probing its mechanism of action against bacteria using a variety of assays and transmission electron microscopy (TEM) imaging, we reveal that BA-3/4-Butyl exerts its antimicrobial activity via membrane permeabilization. Our findings provide insights into how BA-3/4-Butyl compromises bacterial membranes without causing toxicity in its mammalian counterparts. This study advances the understanding of BA-3/4-Butyl's antimicrobial activity and potential mechanisms of action, ultimately aiding the development of similar novel therapeutic agents to help combat antimicrobial resistance.
Collapse
Affiliation(s)
- Colin
C. Tang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Conor Pulliam
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Alimi Abiodun
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Adam Parris
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Andrew Campbell
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Jie Li
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| |
Collapse
|
20
|
Ku J, Hsu J, Li Y, Wu L. Interplay among IL1R1, gut microbiota, and bile acids in metabolic dysfunction-associated steatotic liver disease: a comprehensive review. J Gastroenterol Hepatol 2025; 40:33-40. [PMID: 39343617 PMCID: PMC11771549 DOI: 10.1111/jgh.16750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/07/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent metabolic disorder characterized by hepatic steatosis associated with metabolic abnormalities. Recent research has shed light on the intricate interplay among interleukin-1 receptor 1 (IL1R1), gut microbiota, and bile acids in the pathogenesis of MASLD. This review aims to provide a comprehensive overview of the current understanding of the role of IL1R1, gut microbiota, and bile acids in MASLD, exploring their interrelationships and potential mechanisms. We summarize the evidence supporting the involvement of IL1R1 in inflammation, discuss the influence of gut microbiota on bile acid metabolism and its influence on liver health, and elucidate the bidirectional interactions among IL1R1 signaling, gut microbiota composition, and bile acid homeostasis in MASLD. Furthermore, we highlight emerging therapeutic strategies targeting these interrelated pathways for the management of MASLD.
Collapse
Affiliation(s)
- Jie‐Lun Ku
- School of Medicine, College of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Jia‐Rou Hsu
- Department and Institute of Physiology, College of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yung‐Tsung Li
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipei100Taiwan
| | - Li‐Ling Wu
- Department and Institute of Physiology, College of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Health Innovation CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Microbiota Research CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
21
|
Gurumayum N, Devi MB, Khound P, Bhattacharya A, Sarma H, Khan MR, Devi R. Bioactive fraction of Musa balbisiana seed mitigates D-galactose-induced brain aging via SIRT1/PGC-1α/FoxO3a activation and intestinal barrier dysfunction by modulating gut microbiota and core metabolites. Free Radic Biol Med 2025; 226:43-55. [PMID: 39542187 DOI: 10.1016/j.freeradbiomed.2024.11.016] [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: 06/18/2024] [Revised: 10/29/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Aging is an inevitable biological process, and emerging research has highlighted the potential of dietary and pharmacological interventions to decelerate the trajectory of age-related diseases and prolong the health span. This study evaluates the protective effects of Musa balbisiana seed on healthy aging using D-galactose-induced accelerated aging rats. The results suggested that the bioactive ethyl acetate fraction of Musa balbisiana seed extract (BF) exhibited protective effects against aging-induced oxidative stress by reducing oxidative DNA damage, advanced glycation end-product formation, and malondialdehyde levels while restoring antioxidant and glyoxalase enzyme activities. BF also ameliorated neurodegeneration by decreasing acetylcholinesterase enzyme activity and amyloid beta plaque formation. Histopathological analysis demonstrated the protective effects of BF against brain aging, liver disruption, renal damage, and intestinal barrier dysfunction. BF further restored intestinal permeability by upregulating the tight junctions (zonula occludens 1 and 2, claudin 1,2,3 and 4, and occludin) and mucin (mucin 2 and mucin 5ac) gene expression while downregulating the expression of inflammatory cytokines (IL-1β, IL-6, and TNF-α). BF significantly induced the phosphorylation of FoxO3a proteins and upregulated the gene expression of SIRT1, PGC-1α, and TFAM in the hippocampus. Next-generation sequencing (NGS) of 16s rRNA amplicons of fecal metagenomics DNA and metabolites profiling showed that BF intervention restructured the gut microbiota and altered core metabolites related to cholesterol metabolism. Overall, our findings demonstrated the multifaceted protective effects of Musa balbisiana seed against D-galactose-induced aging.
Collapse
Affiliation(s)
- Nonibala Gurumayum
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati-781035, Assam, India; Department of Zoology, Gauhati University, Guwahati, 781014, India
| | - M Bidyarani Devi
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati-781035, Assam, India; Department of Biotechnology, Gauhati University, Guwahati, 781014, India
| | - Puspanjali Khound
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati-781035, Assam, India; Department of Zoology, Gauhati University, Guwahati, 781014, India
| | - Anupam Bhattacharya
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati-781035, Assam, India
| | - Himangshu Sarma
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati-781035, Assam, India
| | - Mojibur R Khan
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati-781035, Assam, India.
| | - Rajlakshmi Devi
- Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati-781035, Assam, India; Department of Zoology, Gauhati University, Guwahati, 781014, India.
| |
Collapse
|
22
|
Kamath S, Sokolenko E, Collins K, Chan NSL, Mills N, Clark SR, Marques FZ, Joyce P. IUPHAR themed review: The gut microbiome in schizophrenia. Pharmacol Res 2025; 211:107561. [PMID: 39732352 DOI: 10.1016/j.phrs.2024.107561] [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: 11/25/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Gut microbial dysbiosis or altered gut microbial consortium, in schizophrenia suggests a pathogenic role through the gut-brain axis, influencing neuroinflammatory and neurotransmitter pathways critical to psychotic, affective, and cognitive symptoms. Paradoxically, conventional psychotropic interventions may exacerbate this dysbiosis, with antipsychotics, particularly olanzapine, demonstrating profound effects on microbial architecture through disruption of bacterial phyla ratios, diminished taxonomic diversity, and attenuated short-chain fatty acid synthesis. To address these challenges, novel therapeutic strategies targeting the gut microbiome, encompassing probiotic supplementation, prebiotic compounds, faecal microbiota transplantation, and rationalised co-pharmacotherapy, show promise in attenuating antipsychotic-induced metabolic disruptions while enhancing therapeutic efficacy. Harnessing such insights, precision medicine approaches promise to transform antipsychotic prescribing practices by identifying patients at risk of metabolic side effects based on their microbial profiles. This IUPHAR review collates the current literature landscape of the gut-brain axis and its intricate relationship with schizophrenia while advocating for integrating microbiome assessments and therapeutic management. Such a fundamental shift in proposing microbiome-informed psychotropic prescriptions to optimise therapeutic efficacy and reduce adverse metabolic impacts would align antipsychotic treatments with microbiome safety, prioritising 'gut-neutral' or gut-favourable drugs to safeguard long-term patient outcomes in schizophrenia therapy.
Collapse
Affiliation(s)
- Srinivas Kamath
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Elysia Sokolenko
- Discipline of Anatomy and Pathology, School of Biomedicine, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Kate Collins
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Nicole S L Chan
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Natalie Mills
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Scott R Clark
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Francine Z Marques
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Hypertension Research Laboratory, School of Biological Sciences and Victorian Heart Institute, Monash University, Melbourne, VIC, Australia
| | - Paul Joyce
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia.
| |
Collapse
|
23
|
Gautam R, Maan P, Patel AK, Vasudevan S, Arora T. Unveiling the complex interplay between gut microbiota and polycystic ovary syndrome: A narrative review. Clin Nutr 2024; 43:199-208. [PMID: 39481287 DOI: 10.1016/j.clnu.2024.10.028] [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: 04/04/2024] [Revised: 07/11/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND & AIM Polycystic Ovary Syndrome (PCOS) is a complex endocrine disorder that affects women throughout their reproductive age and characterised via polycystic ovaries, hyperandrogenism, and irregular menstruation. There is rising evidence that the pathophysiology of PCOS is significantly affected via the gut microbiota and its metabolic products. METHODS This narrative review synthesizes current literature exploring the relationship between gut microbiota and PCOS. A comprehensive search of electronic databases was conducted to identify relevant studies. Further this review also analysed therapeutic options of probiotics, prebiotics, Fecal Microbiota Transplant (FMT), high fiber and poly phenol rich diet and novel therapeutic agents in treatment of PCOS. RESULTS Emerging evidence suggests alterations in the composition and diversity of gut microbiota in women with PCOS. The current literature showed a complex relationship of gut microbiota, short chain fatty acids (SCFAs) metabolism, intestinal permeability and LPS (Lipid Polysaccharide) metabolism, gut-brain axis and bile acid (BA) pathway within etiology and pathophysiology of PCOS. Additionally, the factors such as diet, lifestyle, genetics, and environmental influences may all contribute to alterations in gut microbiota that could potentially exacerbate or mitigate PCOS symptoms. CONCLUSION The review provides valuable insights into the intricate interplay between the gut and female reproductive health. The present evidence suggested that alterations in diversity and function of the gut microbiota may lead to specific pathogenic changes that lead to development of PCOS. A comprehensive understanding of these microbial dynamics may lead to new therapeutic approaches that target the gut micro biome.
Collapse
Affiliation(s)
- Rohit Gautam
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India
| | - Pratibha Maan
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India
| | - Arbind Kumar Patel
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Sudharsan Vasudevan
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India
| | - Taruna Arora
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India.
| |
Collapse
|
24
|
Chen WB, Hu GA, Dong BC, Sun HY, Lu DZ, Ru MY, Yu YL, Wang H, Wei B. Insights into the modulatory effects of host-gut microbial xanthine co-metabolism on high-fat diet-fed mice. Biochem Pharmacol 2024; 230:116596. [PMID: 39481656 DOI: 10.1016/j.bcp.2024.116596] [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: 03/07/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Gut microbiota-mediated endobiotic and xenobiotic metabolism play crucial roles in disease progression, and drug therapy/toxicity. Our recent study suggested that gut microbiota-mediated xanthine metabolism is correlated with resistance to high-fat diet (HFD)-induced obesity. Here, we explored the role of host-gut microbial xanthine co-metabolism in the prevention and treatment of HFD-induced obesity by orally administration of Bifidobacterium longum, xanthine, and a xanthine oxidase inhibitor (topiroxostat). The findings indicate that xanthine exhibits a significantly protective effect against HFD-induced obesity. While B. longum, xanthine, and topiroxostat did not alleviate the dysbiosis of the weight and glucose metabolism of HFD-induced obesity (DIO) and obesity resistance (DIR) mice. 16S rRNA sequencing analyses revealed that treatments with B. longum significantly altered gut microbiota composition in HFD-fed and DIO mice. Microbial interaction network analysis revealed several Bacteroidetes species, such as Amulumruptor caecigallinarius and Muribaculum intestinale, as keystone taxa that were notably enriched by B. longum. Untargeted metabolomics analysis implied that xanthine might serve as a crucial molecule in regulating body weight, exerting a preventive effect on HFD-induced obesity. This study offers new perspectives on the influence of host-gut microbial xanthine co-metabolism on HFD-fed mice and emphasizes the promising role of xanthine in promoting weight loss.
Collapse
Affiliation(s)
- Wei-Bing Chen
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou 310014, China
| | - Gang-Ao Hu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou 310014, China
| | - Bing-Cheng Dong
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou 310014, China
| | - Huai-Ying Sun
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou 310014, China
| | - Dong-Ze Lu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou 310014, China
| | - Meng-Ying Ru
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yan-Lei Yu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hong Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou 310014, China; Binjiang Institute of Artificial Intelligence, ZJUT, Hangzhou 310056, China.
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou 310014, China; Binjiang Institute of Artificial Intelligence, ZJUT, Hangzhou 310056, China.
| |
Collapse
|
25
|
Duan J, Zhong Q, Luo L, Ning Y, Qi Z, Wang S, Liang K. Metabolomic profiling of human feces and plasma from extrauterine growth restriction infants. Pediatr Res 2024:10.1038/s41390-024-03690-7. [PMID: 39496876 DOI: 10.1038/s41390-024-03690-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/18/2024] [Accepted: 10/04/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Extrauterine growth restriction (EUGR) affects a substantial proportion of preterm infants and may influence both short-term complications and long-term sequelae. While many preterm infants with EUGR are secondary to small for gestational age (SGA) or very low birth weight (VLBW), a subset of EUGR infants do not exhibit these conditions. The purpose of this study is to investigate the metabolic profiles and biomarkers of EUGR infants in the absence of SGA and VLBW. METHODS A total of 100 feces (n = 50) and plasma samples (n = 50) were collected from participants categorized as either EUGR (EUGR group) or non-EUGR (NonEUGR group) in the absence of SGA and VLBW. Metabolites were characterized via UPLC-MS/MS using the Discovery HD4® platform. Data normalization, partial least squares discriminant analysis (PLSDA), and KEGG enrichment analysis of metabolite profiles were performed using the MetaboAnalyst 6.0. RESULTS The clinical characteristics of preterm infants differed significantly between the EUGR and NonEUGR groups at discharge, including length of stay, weight Z-score, weight, height Z-score, height, head circumference, and fat-free mass. The PLSDA model exhibited clustering within groups and separation between groups. A total of 58 and 71 differential metabolites were identified in feces and plasma samples, respectively. They were involved in pathways such as caffeine, galactose, glutathione, cysteine, and methionine metabolisms. In the feces sample, 1-palmitoyl-galactosylglycerol exhibited a significant negative correlation with the growth characteristics of preterm infants, while 1-palmitoyl-2-palmitoleoyl-GPC displayed the opposite pattern. In plasma samples, androsterone glucuronide displayed a significant positive correlation with the growth characteristics of preterm infants, whereas 2-methoxyhydroquinone sulfate generated an opposite pattern. Moreover, 2-oleoylglycerol and sphinganine-1-phosphate exhibited the highest area under the curve in feces and plasma samples, respectively, according to diagnostic ROC curves. CONCLUSION Preterm infants with EUGR, in the absence of SGA and VLBW, exhibit specific clinical characteristics and metabolomic profiles. Sphinganine-1-phosphate and 2-oleoylglycerol may hold promise as diagnostic markers for EUGR in the absence of SGA and VLBW. IMPACT The objective of this study is to identify the differential metabolites in preterm infants with extrauterine growth restriction (EUGR) in the absence of small for gestational age (SGA) or very low birth weight (VLBW). Preterm infants with EUGR without SGA and VLBW exhibit specific clinical characteristics and metabolomic profiles. Sphinganine-1-phosphate and 2-oleoylglycerol emerged as potential diagnostic biomarkers for EUGR. This study enhances our understanding of the metabolomic profile in preterm infants with EUGR without SGA or VLBW. Our findings will offer valuable evidence for improving nutritional management and shedding light on the associated pathophysiological mechanisms of EUGR.
Collapse
Affiliation(s)
- Jiang Duan
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Qinghua Zhong
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Liyan Luo
- Department of Neonatology, Dali Prefecture Maternal and Child Health Care Hospital, Dali, Yunnan Province, China
| | - Yue Ning
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Zhiye Qi
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Sixian Wang
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu, Sichuan Province, China
| | - Kun Liang
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| |
Collapse
|
26
|
Mansour H, Slika H, Nasser SA, Pintus G, Khachab M, Sahebkar A, Eid AH. Flavonoids, gut microbiota and cardiovascular disease: Dynamics and interplay. Pharmacol Res 2024; 209:107452. [PMID: 39383791 DOI: 10.1016/j.phrs.2024.107452] [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: 01/28/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Cardiovascular disease (CVD) remains the leading cause of global morbidity and mortality. Extensive efforts have been invested to explicate mechanisms implicated in the onset and progression of CVD. Besides the usual suspects as risk factors (obesity, diabetes, and others), the gut microbiome has emerged as a prominent and essential factor in the pathogenesis of CVD. With its endocrine-like effects, the microbiome modulates many physiologic processes. As such, it is not surprising that dysbiosis-by generating metabolites, inciting inflammation, and altering secondary bile acid signaling- could predispose to or aggravate CVD. Nevertheless, various natural and synthetic compounds have been shown to modulate the microbiome. Prime among these molecules are flavonoids, which are natural polyphenols mainly present in fruits and vegetables. Accumulating evidence supports the potential of flavonoids in attenuating the development of CVD. The ascribed mechanisms of these compounds appear to involve mitigation of inflammation, alteration of the microbiome composition, enhancement of barrier integrity, induction of reverse cholesterol transport, and activation of farnesoid X receptor signaling. In this review, we critically appraise the methods by which the gut microbiome, despite being essential to the human body, predisposes to CVD. Moreover, we dissect the mechanisms and pathways underlying the cardioprotective effects of flavonoids.
Collapse
Affiliation(s)
- Hadi Mansour
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hasan Slika
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy
| | - Maha Khachab
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Beirut, Lebanon
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
27
|
Yan Q, Xing J, Zou R, Sun M, Zou B, Wang Y, Niu T, Yu T, Huang H, Yang W, Shi C, Yang G, Wang C. LysoPE mediated by respiratory microorganism Aeromicrobium camelliae alleviates H9N2 challenge in mice. Vet Res 2024; 55:136. [PMID: 39390593 PMCID: PMC11468851 DOI: 10.1186/s13567-024-01391-x] [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: 04/08/2024] [Accepted: 08/13/2024] [Indexed: 10/12/2024] Open
Abstract
Influenza remains a severe respiratory illness that poses significant global health threats. Recent studies have identified distinct microbial communities within the respiratory tract, from nostrils to alveoli. This research explores specific anti-influenza respiratory microbes using a mouse model supported by 16S rDNA sequencing and untargeted metabolomics. The study found that transferring respiratory microbes from mice that survived H9N2 influenza to antibiotic-treated mice enhanced infection resistance. Notably, the levels of Aeromicrobium were significantly higher in the surviving mice. Mice pre-treated with antibiotics and then inoculated with Aeromicrobium camelliae showed reduced infection severity, as evidenced by decreased weight loss, higher survival rates, and lower lung viral titres. Metabolomic analysis revealed elevated LysoPE (16:0) levels in mildly infected mice. In vivo and in vitro experiments indicated that LysoPE (16:0) suppresses inducible nitric oxide synthase (INOS) and cyclooxygenase-2 (COX2) expression, enhancing anti-influenza defences. Our findings suggest that Aeromicrobium camelliae could serve as a potential agent for influenza prevention and a prognostic marker for influenza outcomes.
Collapse
Affiliation(s)
- Qingsong Yan
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Junhong Xing
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Ruonan Zou
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Mingjie Sun
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Boshi Zou
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yingjie Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Tianming Niu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Tong Yu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Haibin Huang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wentao Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Chunwei Shi
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| | - Guilian Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
28
|
Luo T, Che Q, Guo Z, Song T, Zhao J, Xu D. Modulatory effects of traditional Chinese medicines on gut microbiota and the microbiota-gut-x axis. Front Pharmacol 2024; 15:1442854. [PMID: 39444598 PMCID: PMC11497133 DOI: 10.3389/fphar.2024.1442854] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
The gut microbiota offers numerous benefits to the human body, including the promotion of nutrient absorption, participation in metabolic processes, and enhancement of immune function. Recent studies have introduced the concept of the gut-organ axis, which encompasses interactions such as the gut-brain axis, gut-liver axis, and gut-lung axis. This concept underscores the complex interplay between gut microbiota and various organs and tissues, including the brain, heart, lungs, liver, kidneys, muscles, and bones. Growing evidence indicates that gut microbiota can influence the onset and progression of multi-organ system diseases through their effects on the gut-organ axis. Traditional Chinese medicine has demonstrated significant efficacy in regulating the gastrointestinal system, leveraging its unique advantages. Considerable advancements have been made in understanding the role of gut microbiota and the gut-organ axis within the mechanisms of action of traditional Chinese medicine. This review aims to elucidate the roles of gut microbiota and the gut-organ axis in human health, explore the potential connections between traditional Chinese medicine and gut microbiota, and examine the therapeutic effects of traditional Chinese medicine on the microbiota-gut-organ axis. Furthermore, the review addresses the limitations and challenges present in current research while proposing potential directions for future investigations in this area.
Collapse
Affiliation(s)
- Tingting Luo
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, China
| | - Qingya Che
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, China
| | - Ziyi Guo
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, China
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Tingxia Song
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, China
| | - Juanjuan Zhao
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Delin Xu
- Department of Medical Instrumental Analysis, Zunyi Medical University, Zunyi, China
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
29
|
Kazemian N, Pakpour S. Understanding the impact of the gut microbiome on opioid use disorder: Pathways, mechanisms, and treatment insights. Microb Biotechnol 2024; 17:e70030. [PMID: 39388360 PMCID: PMC11466222 DOI: 10.1111/1751-7915.70030] [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: 05/14/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
The widespread use of opioids for chronic pain management not only poses a significant public health issue but also contributes to the risk of tolerance, dependence, and addiction, leading to opioid use disorder (OUD), which affects millions globally each year. Recent research has highlighted a potential bidirectional relationship between the gut microbiome and OUD. This emerging perspective is critical, especially as the opioid epidemic intensifies, emphasizing the need to investigate how OUD may alter gut microbiome dynamics and vice versa. Understanding these interactions could reveal new insights into the mechanisms of addiction and tolerance, as well as provide novel approaches for managing and potentially mitigating OUD impacts. This comprehensive review explores the intricate bidirectional link through the gut-brain axis, focusing on how opiates influence microbial composition, functional changes, and gut mucosal integrity. By synthesizing current findings, the review aims to inspire new strategies to combat the opioid crisis and leverage microbiome-centred interventions for preventing and treating OUD.
Collapse
Affiliation(s)
- Negin Kazemian
- School of EngineeringUniversity of British ColumbiaKelownaBritish ColumbiaCanada
| | - Sepideh Pakpour
- School of EngineeringUniversity of British ColumbiaKelownaBritish ColumbiaCanada
| |
Collapse
|
30
|
González A, Fullaondo A, Odriozola I, Odriozola A. Microbiota and other detrimental metabolites in colorectal cancer. ADVANCES IN GENETICS 2024; 112:309-365. [PMID: 39396839 DOI: 10.1016/bs.adgen.2024.08.006] [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: 10/15/2024]
Abstract
Increasing scientific evidence demonstrates that gut microbiota plays an essential role in the onset and development of Colorectal cancer (CRC). However, the mechanisms by which these microorganisms contribute to cancer development are complex and far from completely clarified. Specifically, the impact of gut microbiota-derived metabolites on CRC is undeniable, exerting both protective and detrimental effects. This paper examines the effects and mechanisms by which important bacterial metabolites exert detrimental effects associated with increased risk of CRC. Metabolites considered include heterocyclic amines and polycyclic aromatic hydrocarbons, heme iron, secondary bile acids, ethanol, and aromatic amines. It is necessary to delve deeper into the mechanisms of action of these metabolites in CRC and identify the microbiota members involved in their production. Furthermore, since diet is the main factor capable of modifying the intestinal microbiota, conducting studies that include detailed descriptions of dietary interventions is crucial. All this knowledge is essential for developing precision nutrition strategies to optimise a protective intestinal microbiota against CRC.
Collapse
Affiliation(s)
- Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Asier Fullaondo
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Iñaki Odriozola
- Health Department of Basque Government, Donostia-San Sebastián, Spain
| | - Adrian Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain.
| |
Collapse
|
31
|
Ferraz-Bannitz R, Ozturk B, Cummings C, Efthymiou V, Casanova Querol P, Poulos L, Wang H, Navarrete V, Saeed H, Mulla CM, Pan H, Dreyfuss JM, Simonson DC, Sandoval DA, Patti ME. Postprandial metabolomics analysis reveals disordered serotonin metabolism in post-bariatric hypoglycemia. J Clin Invest 2024; 134:e180157. [PMID: 39264731 PMCID: PMC11527454 DOI: 10.1172/jci180157] [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: 02/21/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUNDBariatric surgery is a potent therapeutic approach for obesity and type 2 diabetes but can be complicated by post-bariatric hypoglycemia (PBH). PBH typically occurs 1-3 hours after meals, in association with exaggerated postprandial levels of incretins and insulin.METHODSTo identify mediators of disordered metabolism in PBH, we analyzed the plasma metabolome in the fasting state and 30 and 120 minutes after mixed meal in 3 groups: PBH (n = 13), asymptomatic post-Roux-en-Y gastric bypass (post-RYGB) (n = 10), and nonsurgical controls (n = 8).RESULTSIn the fasting state, multiple tricarboxylic acid cycle intermediates and the ketone β-hydroxybutyrate were increased by 30%-80% in PBH versus asymptomatic. Conversely, multiple amino acids (branched-chain amino acids, tryptophan) and polyunsaturated lipids were reduced by 20%-50% in PBH versus asymptomatic. Tryptophan-related metabolites, including kynurenate, xanthurenate, and serotonin, were reduced 2- to 10-fold in PBH in the fasting state. Postprandially, plasma serotonin was uniquely increased 1.9-fold in PBH versus asymptomatic post-RYGB. In mice, serotonin administration lowered glucose and increased plasma insulin and GLP-1. Moreover, serotonin-induced hypoglycemia in mice was blocked by the nonspecific serotonin receptor antagonist cyproheptadine and the specific serotonin receptor 2 antagonist ketanserin.CONCLUSIONTogether these data suggest that increased postprandial serotonin may contribute to the pathophysiology of PBH and provide a potential therapeutic target.FUNDINGNational Institutes of Health (NIH) grant R01-DK121995, NIH grant P30-DK036836 (Diabetes Research Center grant, Joslin Diabetes Center), and Fundação de Amparo à Pesquisa do Estado de São Paulo grant 2018/22111-2.
Collapse
Affiliation(s)
- Rafael Ferraz-Bannitz
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Berkcan Ozturk
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Cameron Cummings
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Vissarion Efthymiou
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Pilar Casanova Querol
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Lindsay Poulos
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Hanna Wang
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Valerie Navarrete
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Hamayle Saeed
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher M. Mulla
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Hui Pan
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Jonathan M. Dreyfuss
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Donald C. Simonson
- Harvard Medical School, Boston, Massachusetts, USA
- Divsion of Endocrinology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Darleen A. Sandoval
- Section of Nutrition, Department of Pediatrics, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mary-Elizabeth Patti
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
32
|
DeMichele E, Buret AG, Taylor CT. Hypoxia-inducible factor-driven glycolytic adaptations in host-microbe interactions. Pflugers Arch 2024; 476:1353-1368. [PMID: 38570355 PMCID: PMC11310250 DOI: 10.1007/s00424-024-02953-w] [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: 02/06/2024] [Revised: 02/07/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Mammalian cells utilize glucose as a primary carbon source to produce energy for most cellular functions. However, the bioenergetic homeostasis of cells can be perturbed by environmental alterations, such as changes in oxygen levels which can be associated with bacterial infection. Reduction in oxygen availability leads to a state of hypoxia, inducing numerous cellular responses that aim to combat this stress. Importantly, hypoxia strongly augments cellular glycolysis in most cell types to compensate for the loss of aerobic respiration. Understanding how this host cell metabolic adaptation to hypoxia impacts the course of bacterial infection will identify new anti-microbial targets. This review will highlight developments in our understanding of glycolytic substrate channeling and spatiotemporal enzymatic organization in response to hypoxia, shedding light on the integral role of the hypoxia-inducible factor (HIF) during host-pathogen interactions. Furthermore, the ability of intracellular and extracellular bacteria (pathogens and commensals alike) to modulate host cellular glucose metabolism will be discussed.
Collapse
Affiliation(s)
- Emily DeMichele
- School of Medicine and Systems Biology Ireland, The Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Andre G Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Cormac T Taylor
- School of Medicine and Systems Biology Ireland, The Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
33
|
Severino A, Tohumcu E, Tamai L, Dargenio P, Porcari S, Rondinella D, Venturini I, Maida M, Gasbarrini A, Cammarota G, Ianiro G. The microbiome-driven impact of western diet in the development of noncommunicable chronic disorders. Best Pract Res Clin Gastroenterol 2024; 72:101923. [PMID: 39645277 DOI: 10.1016/j.bpg.2024.101923] [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: 04/14/2024] [Accepted: 05/02/2024] [Indexed: 07/23/2024]
Abstract
Noncommunicable chronic disorders (NCDs) are multifactorial disorders that share a state of chronic, low-grade inflammation together with an imbalance of gut microbiota. NCDs are becoming increasingly prevalent worldwide, and mainly in Western countries, with a significant impact on global health. Societal changes, together with the widespread diffusion of modern agricultural methods and food processing, have led to a significant shift in dietary habits over the past century, with an increased diffusion of the Western diet (WD). WD includes foods high in saturated fat, refined sugars, salt, sweeteners, and low in fiber, and is characterized by overeating, frequent snacking, and a prolonged postprandial state. An increasing body of evidence supports the association between the diffusion of WD and the rising prevalence of NCDs. WD also negatively affects both gut microbiota and the immune system by driving to microbial alterations, gut barrier dysfunction, increased intestinal permeability, and leakage of harmful bacterial metabolites into the bloodstream, with consequent contribution to the development of systemic low-grade inflammation. In this review article we aim to dissect the role of gut microbiota imbalance and gut barrier impairment in mediating the detrimental effects of WD on the development of NCDs, and to identify potential therapeutic strategies.
Collapse
Affiliation(s)
- Andrea Severino
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy.
| | - Ege Tohumcu
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Luca Tamai
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Pasquale Dargenio
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Serena Porcari
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Debora Rondinella
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Irene Venturini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Marcello Maida
- Department of Medicine and Surgery, University of Enna 'Kore', Enna, Italy; Gastroenterology Unit, Umberto I Hospital, Enna, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
34
|
Compare D, Sgamato C, Rocco A, Coccoli P, Ambrosio C, Nardone G. The Leaky Gut and Human Diseases: "Can't Fill the Cup if You Don't Plug the Holes First". Dig Dis 2024; 42:548-566. [PMID: 39047703 DOI: 10.1159/000540379] [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: 03/25/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND The gut barrier is a sophisticated and dynamic system that forms the frontline defense between the external environment and the body's internal milieu and includes various structural and functional components engaged not only in digestion and nutrient absorption but also in immune regulation and overall health maintenance. SUMMARY When one or more components of the intestinal barrier lose their structure and escape their function, this may result in a leaky gut. Mounting evidence emphasizes the crucial role of the gut microbiome in preserving the integrity of the gut barrier and provides insights into the pathophysiological implications of conditions related to leaky gut in humans. Assessment of intestinal permeability has evolved from invasive techniques to noninvasive biomarkers, but challenges remain in achieving consensus about the best testing methods and their accuracy. Research on the modulation of gut permeability is just starting, and although no medical guidelines for the treatment of leaky gut syndrome are available, several treatment strategies are under investigation with promising results. KEY MESSAGES This review discusses the composition of the intestinal barrier, the pathophysiology of the leaky gut and its implications on human health, the measurement of intestinal permeability, and the therapeutic strategies to restore gut barrier integrity.
Collapse
Affiliation(s)
- Debora Compare
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Costantino Sgamato
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Alba Rocco
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Pietro Coccoli
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Carmen Ambrosio
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| | - Gerardo Nardone
- Department of Clinical Medicine and Surgery, Gastroenterology, University Federico II of Naples, Naples, Italy
| |
Collapse
|
35
|
Karakus E, Proksch AL, Moritz A, Geyer J. Quantitative bile acid profiling in healthy adult dogs and pups from serum, plasma, urine, and feces using LC-MS/MS. Front Vet Sci 2024; 11:1380920. [PMID: 38948668 PMCID: PMC11211631 DOI: 10.3389/fvets.2024.1380920] [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: 02/02/2024] [Accepted: 04/29/2024] [Indexed: 07/02/2024] Open
Abstract
Synthesis and secretion of bile acids (BA) is a key physiological function of the liver. In pathological conditions like portosystemic shunt, hepatic insufficiency, hepatitis, or cirrhosis BA metabolism and secretion are disturbed. Quantification of total serum BA is an established diagnostic method to assess the general liver function and allows early detection of abnormalities, liver disease progression and guidance of treatment decisions. To date, data on comparative BA profiles in dogs are limited. However, BA profiles might be even better diagnostic parameters than total BA concentrations. On this background, the present study analyzed and compared individual BA profiles in serum, plasma, urine, and feces of 10 healthy pups and 40 adult healthy dogs using ultra-high performance liquid chromatography coupled to electrospray ionization mass spectrometry. Sample preparation was performed by solid-phase extraction for serum, plasma, and urine samples or by protein precipitation with methanol for the feces samples. For each dog, 22 different BA, including unconjugated BA and their glycine and taurine conjugates, were analyzed. In general, there was a great interindividual variation for the concentrations of single BA, mostly exemplified by the fact that cholic acid (CA) was by far the most prominent BA in blood and urine samples of some of the dogs (adults and pups), while in others, CA was under the detection limit. There were no significant age-related differences in the BA profiles, but pups showed generally lower absolute BA concentrations in serum, plasma, and urine. Taurine-conjugated BA were predominant in the serum and plasma of both pups (68%) and adults (74-75%), while unconjugated BA were predominant in the urine and feces of pups (64 and 95%, respectively) and adults (68 and 99%, respectively). The primary BA chenodeoxycholic acid and taurocholic acid and the secondary BA deoxycholic acid and lithocholic acid were the most robust analytes for potential diagnostic purpose. In conclusion, this study reports simultaneous BA profiling in dog serum, plasma, urine, and feces and provides valuable diagnostic data for subsequent clinical studies in dogs with different kinds of liver diseases.
Collapse
Affiliation(s)
- Emre Karakus
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Anna-Lena Proksch
- Clinic of Small Animals—Internal Medicine, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Andreas Moritz
- Clinic of Small Animals—Internal Medicine, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Joachim Geyer
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
36
|
Du X, Liu M, Trevisi E, Ju L, Yang Y, Gao W, Song Y, Lei L, Zolzaya M, Li X, Fang Z, Liu G. Expression of hepatic genes involved in bile acid metabolism in dairy cows with fatty liver. J Dairy Sci 2024:S0022-0302(24)00833-6. [PMID: 38825110 DOI: 10.3168/jds.2023-24485] [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: 11/29/2023] [Accepted: 04/28/2024] [Indexed: 06/04/2024]
Abstract
Bile acids are cholesterol-derived molecules that are primarily produced in the liver. In nonruminants with fatty liver, overproduction of bile acids is associated with liver injury. During the transition period, fatty liver is a metabolic disorder that can affect up to 50% of high-producing dairy cows. The purpose of this study was to provide a comprehensive evaluation on hepatic bile acid metabolism in dairy cows with fatty liver by assessing expression changes of genes involved in bile acid synthesis, export and uptake. The serum activities of aspartate aminotransferase, alanine aminotransferase and glutamate dehydrogenase and concentration of total bile acids were all greater, whereas serum concentration of total cholesterol was lower in cows with fatty liver than in healthy cows. Content of total bile acids was higher but total cholesterol was slightly lower in liver tissues from fatty liver cows than from healthy cows. The hepatic mRNA abundance of cholesterol 7a-hydroxylase (CYP7A1), hydroxy-delta-5-steroid dehydrogenase, 3 β- and steroid delta-isomerase 7 (HSD3B7) and sterol 12α-hydroxylase (CYP8B1), enzymes involved in the classic pathway of bile acid synthesis, was higher in fatty liver cows than in healthy cows. Compared with healthy cows, the hepatic mRNA abundance of alternative bile acid synthesis pathway-related genes sterol 27-hydroxylase (CYP27A1) and oxysterol 7α-hydroxylase (CYP7B1) did not differ in cows with fatty liver. The protein and mRNA abundance of bile acid transporter bile salt efflux pump (BSEP) were lower in the liver of dairy cow with fatty liver. Compared with healthy cows, the hepatic mRNA abundance of bile acid transporters solute carrier family 51 subunit α (SLC51A), ATP binding cassette subfamily C member 1 (ABCC1) and 3 (ABCC3) was greater in cows with fatty liver, whereas the solute carrier family 51 subunit β (SLC51B) did not differ. The expression of genes involved in bile acid uptake, including solute carrier family 10 member 1 (NTCP), solute carrier organic anion transporter family member 1A2 (SLCO1A2) and 2B1 (SLCO2B1) was upregulated in dairy cows with fatty liver. Furthermore, the hepatic protein and mRNA abundance of bile acid metabolism regulators farnesoid X receptor (FXR) and small heterodimer partner (SHP) were lower in cows with fatty liver than in healthy cows. Overall, these data suggest that inhibition of FXR signaling pathway may lead to the increased bile acid synthesis and uptake and decreased secretion of bile acids from hepatocytes to the bile, which elevates hepatic bile acids content in dairy cows with fatty liver. As the hepatotoxicity of bile acids has been demonstrated on nonruminant hepatocytes, it is likely that the liver injury is induced by increased hepatic bile acids content in dairy cows with fatty liver.
Collapse
Affiliation(s)
- Xiliang Du
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Mingchao Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, 071001, Hebei, China
| | - Erminio Trevisi
- Department of Animal Sciences, Food and Nutrition, Faculty of Agriculture, Food and Environmental Science, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - Lingxue Ju
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuting Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenwen Gao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuxiang Song
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lin Lei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Majigsuren Zolzaya
- Institute of Veterinary Medicine, Mongolian Mongolian University of Life Sciences (MULS)
| | - Xinwei Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zhiyuan Fang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Guowen Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
37
|
Park D, Kim HS, Kim JH. Effect of Pre-Antibiotic Use Before First Stroke Incidence on Recurrence and Mortality: A Longitudinal Study Using the Korean National Health Insurance Service Database. Int J Gen Med 2024; 17:1625-1633. [PMID: 38706744 PMCID: PMC11068048 DOI: 10.2147/ijgm.s456925] [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: 12/27/2023] [Accepted: 04/23/2024] [Indexed: 05/07/2024] Open
Abstract
Purpose Clinical studies on dysbiosis and stroke outcomes has been insufficient to establish clear evidence. This study aimed to investigate the effects of pre-antibiotic use before a stroke event on secondary outcomes using a longitudinal population-level database. Patients and Methods This retrospective cohort study included adults aged 55 years or older diagnosed with acute ischemic stroke (AIS) and acute hemorrhagic stroke (AHS) between 2004 and 2007. Patients were followed-up until the end of 2019, and the target outcomes were secondary AIS, AHS, and all-cause mortality. Multivariable Cox regression analyses were applied, and we adjusted covariates such as age, sex, socioeconomic status, hypertension, diabetes, and dyslipidemia. Pre-antibiotic use was identified from 7 days to 1 year before the acute stroke event. Results We included 159,181 patients with AIS (AIS group) and 49,077 patients with AHS (AHS group). Pre-antibiotic use significantly increased the risk of secondary AIS in the AIS group (adjusted hazard ratio [aHR], 1.03; 95% confidence interval [CI], 1.01-1.05; p = 0.009) and secondary AHS in the AHS group (aHR, 1.08; 95% CI, 1.03-1.12; p <0.001). Furthermore, pre-antibiotic use in the AIS group was associated with a lower risk of mortality (aHR, 0.95; 95% CI, 0.94-0.96; p <0.001). Conclusion Our population-based longitudinal study revealed that pre-antibiotic use was associated with a higher risk of secondary stroke and a lower risk of mortality in the AIS and AHS groups. Further studies are needed to understand the relationship between dysbiosis and stroke outcomes.
Collapse
Affiliation(s)
- Dougho Park
- Medical Research Institute, Pohang Stroke and Spine Hospital, Pohang, Republic of Korea
- Department of Medical Science and Engineering, School of Convergence Science and Technology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Hyoung Seop Kim
- Department of Physical Medicine and Rehabilitation, National Health Insurance Service Ilsan Hospital, Goyang, Republic of Korea
| | - Jong Hun Kim
- Department of Neurology, National Health Insurance Service Ilsan Hospital, Goyang, Republic of Korea
| |
Collapse
|
38
|
Li Y, Xu H, Zhou L, Zhang Y, Yu W, Li S, Gao J. Bifidobacterium breve Protects the Intestinal Epithelium and Mitigates Inflammation in Colitis via Regulating the Gut Microbiota-Cholic Acid Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:3572-3583. [PMID: 38334304 DOI: 10.1021/acs.jafc.3c08527] [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: 02/10/2024]
Abstract
In this study, we aimed to explore the protective effects of Bifidobacterium in colitis mice and the potential mechanisms. Results showed that Bifidobacterium breve (B. breve) effectively colonized the intestinal tract and alleviated colitis symptoms by reducing the disease activity index. Moreover, B. breve mitigated intestinal epithelial cell damage, inhibited the pro-inflammatory factors, and upregulated tight junction (TJ)-proteins. Gut microbiota and metabolome analysis found that B. breve boosted bile acid-regulating genera (such as Bifidobacterium and Clostridium sensu stricto 1), which promoted bile acid deconjugation in the intestine. Notably, cholic acid (CA) was closely associated with the expression levels of inflammatory factors and TJ-proteins (p < 0.05). Our in vitro cell experiments further confirmed that CA (20.24 ± 4.53 pg/mL) contributed to the inhibition of lipopolysaccharide-induced tumor necrosis factor-α expression (49.32 ± 5.27 pg/mL) and enhanced the expression of TJ-proteins (Occludin and Claudin-1) and MUC2. This study suggested that B. breve could be a probiotic candidate for use in infant foods.
Collapse
Affiliation(s)
- Yaqian Li
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Hongtao Xu
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Liuyang Zhou
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Yuwei Zhang
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Wenqing Yu
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Shubo Li
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Jie Gao
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| |
Collapse
|
39
|
Wang D, Ye A, Jiang N. The role of bacteria in gallstone formation. Folia Microbiol (Praha) 2024; 69:33-40. [PMID: 38252338 DOI: 10.1007/s12223-024-01131-w] [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: 06/16/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
Gallstones are a prevalent biliary system disorder that is particularly common in women. They can lead to various complications, such as biliary colic, infection, cholecystitis, and even gallbladder cancer. However, the etiology of gallstones remains incompletely understood. The significant role of bacteria in gallstone formation has been demonstrated in recent studies. Certain bacteria not only influence bile composition and the gallbladder environment but also actively participate in stone formation by producing enzymes such as β-glucuronidase and mucus. Therefore, this review aimed to analyze the mechanisms involving the types and quantities of bacteria involved in gallstone formation, providing valuable references for understanding the etiology and clinical treatment of gallstones.
Collapse
Affiliation(s)
- Danfeng Wang
- Health Management (Physical Examination) Section of Hubei Third People's Hospital Affiliated to Jianghan University, Wuhan, 430034, People's Republic of China
| | - Aihui Ye
- Wuchang District Shouyilu Street Community Health Service Center, Wuhan, 430061, People's Republic of China
| | - Ni Jiang
- Health Management (Physical Examination) Section of Hubei Third People's Hospital Affiliated to Jianghan University, Wuhan, 430034, People's Republic of China.
| |
Collapse
|
40
|
Yang J, Ma G, Wang K, Yang H, Jiang S, Fan Q, Zhou X, Guo G, Han Y. Causal associations between gut microbiota and Cholestatic liver diseases: a Mendelian randomization study. Front Med (Lausanne) 2024; 11:1342119. [PMID: 38327703 PMCID: PMC10847275 DOI: 10.3389/fmed.2024.1342119] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024] Open
Abstract
Background The etiological factors of Cholestatic Liver Diseases especially primary sclerosing cholangitis (PSC) and primary biliary cholangitis (PBC) are not fully illustrated. It has been reported in previous observational studies that gut microbiota are associated with cholestatic liver diseases. However, there is uncertainty regarding the causality of this association. By using Mendelian randomization, this study aimed to examine the causal impact of gut microbiota on cholestatic liver diseases. Methods From large-scale genome-wide association studies, genetic instruments for each gut microbiota taxa as well as primary biliary cholangitis and primary sclerosing cholangitis were developed. Subsequently, we conducted a two-sample Mendelian randomization analysis, supplemented by multiple post hoc sensitivity analyses. Additionally, we performed reverse MR analyses to investigate the possibility of the reverse causal association. Result This two-sample MR study indicated that the order Bacillales, family Peptostreptococcaceae, family Ruminococcaceae, genus Anaerotruncu was associated with a decreased risk of developing PBC, and that order Selenomonadales, family Bifidobacteriaceae may be factors that increase the risk of PBC. On the other hand, we also identified order Selenomonadales, family Rhodospirillaceae, and genus RuminococcaceaeUCG013 were positively associated with PSC. The order Actinomycetales, family Actinomycetaceae, genus Actinomyces, genus Alloprevotella, genus Barnesiella, and genus Peptococcus were found negative associations with the risk of PSC. The reverse MR analysis demonstrated no statistically significant relationship between PBC, PSC and these specific gut microbial taxa. Conclusion Our findings offered novel evidence that the abundance of particular bacteria contributes to the risk of PBC and PSC, which may contribute to more effective approaches to PBC and PSC therapy and prevention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Guanya Guo
- Department of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Ying Han
- Department of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
41
|
Liao SF, Ji F, Fan P, Denryter K. Swine Gastrointestinal Microbiota and the Effects of Dietary Amino Acids on Its Composition and Metabolism. Int J Mol Sci 2024; 25:1237. [PMID: 38279233 PMCID: PMC10816286 DOI: 10.3390/ijms25021237] [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: 11/05/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 01/28/2024] Open
Abstract
Many researchers consider gut microbiota (trillions of microorganisms) an endogenous organ of its animal host, which confers a vast genetic diversity in providing the host with essential biological functions. Particularly, the gut microbiota regulates not only gut tissue structure but also gut health and gut functionality. This paper first summarized those common bacterial species (dominated by the Firmicutes, Bacteroidota, and Proteobacteria phyla) in swine gut and then briefly discussed their roles in swine nutrition and health, which include roles in nutrient metabolism, pathogen exclusion, and immunity modulation. Secondly, the current knowledge on how dietary nutrients and feed additives affect the gut bacterial composition and nutrient metabolism in pigs was discussed. Finally, how dietary amino acids affect the relative abundances and metabolism of bacteria in the swine gut was reviewed. Tryptophan supplementation promotes the growth of beneficial bacteria and suppresses pathogens, while arginine metabolism affects nitrogen recycling, impacting gut immune response and health. Glutamate and glutamine supplementations elevate the levels of beneficial bacteria and mitigate pathogenic ones. It was concluded that nutritional strategies to manipulate gut microbial ecosystems are useful measures to optimize gut health and gut functions. For example, providing pigs with nutrients that promote the growth of Lactobacillus and Bifidobacterium can lead to better gut health and growth performance, especially when dietary protein is limited. Further research to establish the mechanistic cause-and-effect relationships between amino acids and the dynamics of gut microbiota will allow swine producers to reap the greatest return on their feed investment.
Collapse
Affiliation(s)
- Shengfa F. Liao
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA; (P.F.)
| | - Feng Ji
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China;
| | - Peixin Fan
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA; (P.F.)
| | - Kristin Denryter
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39762, USA; (P.F.)
| |
Collapse
|
42
|
Qi H, Wang R, Wang C, Wang R, Shen J, Fang H, Zhang J. Integrated Microbiome and Metabolomics Analysis of the Effects of Dietary Supplementation with Corn-Steep-Liquor-Derived Candida utilis Feed on Black Pigs. Animals (Basel) 2024; 14:306. [PMID: 38254475 PMCID: PMC10812819 DOI: 10.3390/ani14020306] [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: 11/08/2023] [Revised: 12/27/2023] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
In this experiment, glucose master liquor and corn steep liquor were used as carbon and nitrogen sources, and Candida utilis was used as a strain to ferment yeast feed. The OD value and number of yeast cells were used as response values to optimize the medium components of the yeast feed through a response surface methodology. The optimal medium components were a glucose master liquor concentration of 8.3%, a corn steep liquor concentration of 1.2%, and a KH2PO4 concentration of 0.14%. Under this condition of fermentation, the OD value was 0.670 and the number of yeast cells was 2.72 × 108/mL. Then, we fed Candida utilis feed to Dongliao black piglets, and the effects of the yeast feed on the piglets' growth performance, fecal microbiota, and plasma metabolic levels were investigated through 16S rDNA sequencing and metabolomics. In total, 120 black piglets with an average initial weight of 6.90 ± 1.28 kg were randomly divided into two groups. One group was fed the basic diet (the CON group), and the other was supplemented with 2.5% Candida utilis add to the basic diet (the 2.5% CU group). After a pre-feeding period, the formal experiments were performed for 21 days. The results showed that the addition of Candida utilis to the diet did not affect growth performance compared with the control group. Meanwhile, no significant differences were observed in the serum biochemical indices. However, piglets in the 2.5% CU group had a significantly altered fecal microbiota, with an increased abundance of Clostridium_sensu_stricto_1, Lactobacillus, and Muribaculaceae_unclassified. Regarding the plasma metabolome, the 12 differential metabolites detected were mainly enriched in the histidine, tryptophan, primary bile acid, and caffeine metabolic pathways. Regarding the integrated microbiome-metabolome analysis, differential metabolites correlated with fecal flora to variable degrees, but most of them were beneficial bacteria of Firmicutes. Collectively, dietary Candida utilis feed had no adverse effect on growth performance; however, it played an important role in regulating fecal flora and maintaining metabolic levels.
Collapse
Affiliation(s)
| | | | - Chuanqi Wang
- College of Animal Sciences, Jilin University, Changchun 130062, China
| | | | | | | | - Jing Zhang
- College of Animal Sciences, Jilin University, Changchun 130062, China
| |
Collapse
|
43
|
Khan R, Di Gesù CM, Lee J, McCullough LD. The contribution of age-related changes in the gut-brain axis to neurological disorders. Gut Microbes 2024; 16:2302801. [PMID: 38237031 PMCID: PMC10798364 DOI: 10.1080/19490976.2024.2302801] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
Trillions of microbes live symbiotically in the host, specifically in mucosal tissues such as the gut. Recent advances in metagenomics and metabolomics have revealed that the gut microbiota plays a critical role in the regulation of host immunity and metabolism, communicating through bidirectional interactions in the microbiota-gut-brain axis (MGBA). The gut microbiota regulates both gut and systemic immunity and contributes to the neurodevelopment and behaviors of the host. With aging, the composition of the microbiota changes, and emerging studies have linked these shifts in microbial populations to age-related neurological diseases (NDs). Preclinical studies have demonstrated that gut microbiota-targeted therapies can improve behavioral outcomes in the host by modulating microbial, metabolomic, and immunological profiles. In this review, we discuss the pathways of brain-to-gut or gut-to-brain signaling and summarize the role of gut microbiota and microbial metabolites across the lifespan and in disease. We highlight recent studies investigating 1) microbial changes with aging; 2) how aging of the maternal microbiome can affect offspring health; and 3) the contribution of the microbiome to both chronic age-related diseases (e.g., Parkinson's disease, Alzheimer's disease and cerebral amyloidosis), and acute brain injury, including ischemic stroke and traumatic brain injury.
Collapse
Affiliation(s)
- Romeesa Khan
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Claudia M. Di Gesù
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Juneyoung Lee
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Louise D. McCullough
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
44
|
Wu JF, Tseng PH, Chang HH, Chiang CM, Lin WH, Hsu WM, Chang MH. The prevalence and impact of small intestine bacterial overgrowth in biliary atresia patients. Pediatr Res 2024; 95:302-307. [PMID: 37726543 DOI: 10.1038/s41390-023-02818-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/04/2023] [Accepted: 08/30/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Acute cholangitis is an ominous complication in biliary atresia (BA) patients. We investigated the prevalence of small intestine bacterial overgrowth (SIBO) in BA patients and its role in predicting acute cholangitis. METHODS There are 69 BA patients with native liver recruited into this study prospectively. They received hydrogen and methane-based breath testing (HMBT) to detect SIBO after recruitment and were followed prospectively in our institute. RESULTS There are 16 (23.19%) subjects detected to have SIBO by HMBT. BA subjects with SIBO were noted to have higher serum alanine aminotransferase levels than others without SIBO (P = 0.03). The risk of acute cholangitis is significantly higher in BA patients with SIBO than in others without SIBO (62.50% vs. 15.09%, P < 0.001). The logistic regression analysis demonstrated that BA subjects with SIBO have a higher risk of acute cholangitis than others without SIBO (odds ratio = 9.38, P = 0.001). Cox's proportional hazard analysis further confirmed the phenomena in survival analysis (hazard ratio = 6.43, P < 0.001). CONCLUSIONS The prevalence of SIBO in BA patients is 23.19% in this study. The presence of SIBO is associated with the occurrence of acute cholangitis in BA patients. IMPACT What is the key message of your article? Acute cholangitis is common in BA, and is associated with SIBO after hepatoportoenterostomy in this study. What does it add to the existing literature? This study demonstrated that SIBO is common in BA after hepatoportoenterostomy, and is predictive of acute cholangitis and elevated serum ALT levels in BA. What is the impact? This prospective cohort study provides data regarding the significance of SIBO on the risk of acute cholangitis in BA patients.
Collapse
Affiliation(s)
- Jia-Feng Wu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Ping-Huei Tseng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsiu-Hao Chang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Che-Ming Chiang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Hsi Lin
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Mei-Hwei Chang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
45
|
Emencheta SC, Olovo CV, Eze OC, Kalu CF, Berebon DP, Onuigbo EB, Vila MMDC, Balcão VM, Attama AA. The Role of Bacteriophages in the Gut Microbiota: Implications for Human Health. Pharmaceutics 2023; 15:2416. [PMID: 37896176 PMCID: PMC10609668 DOI: 10.3390/pharmaceutics15102416] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/18/2023] [Accepted: 09/30/2023] [Indexed: 10/29/2023] Open
Abstract
Bacteriophages (phages) are nano-sized viruses characterized by their inherent ability to live off bacteria. They utilize diverse mechanisms to absorb and gain entry into the bacterial cell wall via the release of viral genetic material, which uses the replication mechanisms of the host bacteria to produce and release daughter progeny virions that attack the surrounding host cells. They possess specific characteristics, including specificity for particular or closely related bacterial species. They have many applications, including as potential alternatives to antibiotics against multi-resistant bacterial pathogens and as control agents in bacteria-contaminated environments. They are ubiquitously abundant in nature and have diverse biota, including in the gut. Gut microbiota describes the community and interactions of microorganisms within the intestine. As with bacteria, parasitic bacteriophages constantly interact with the host bacterial cells within the gut system and have obvious implications for human health. However, it is imperative to understand these interactions as they open up possible applicable techniques to control gut-implicated bacterial diseases. Thus, this review aims to explore the interactions of bacteriophages with bacterial communities in the gut and their current and potential impacts on human health.
Collapse
Affiliation(s)
- Stephen C. Emencheta
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka 410001, Nigeria; (S.C.E.); (O.C.E.); (C.F.K.); (E.B.O.)
- VBlab—Laboratory of Bacterial Viruses, University of Sorocaba, Sorocaba 18023-000, Brazil; (M.M.D.C.V.); (V.M.B.)
| | - Chinasa V. Olovo
- Department of Microbiology, University of Nigeria, Nsukka 410001, Nigeria;
| | - Osita C. Eze
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka 410001, Nigeria; (S.C.E.); (O.C.E.); (C.F.K.); (E.B.O.)
| | - Chisom F. Kalu
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka 410001, Nigeria; (S.C.E.); (O.C.E.); (C.F.K.); (E.B.O.)
| | - Dinebari P. Berebon
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka 410001, Nigeria; (S.C.E.); (O.C.E.); (C.F.K.); (E.B.O.)
| | - Ebele B. Onuigbo
- Department of Pharmaceutical Microbiology and Biotechnology, University of Nigeria, Nsukka 410001, Nigeria; (S.C.E.); (O.C.E.); (C.F.K.); (E.B.O.)
| | - Marta M. D. C. Vila
- VBlab—Laboratory of Bacterial Viruses, University of Sorocaba, Sorocaba 18023-000, Brazil; (M.M.D.C.V.); (V.M.B.)
| | - Victor M. Balcão
- VBlab—Laboratory of Bacterial Viruses, University of Sorocaba, Sorocaba 18023-000, Brazil; (M.M.D.C.V.); (V.M.B.)
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, P-3810-193 Aveiro, Portugal
| | - Anthony A. Attama
- Department of Pharmaceutics, University of Nigeria, Nsukka 410001, Nigeria
- Institute for Drug-Herbal Medicine-Excipient Research and Development, University of Nigeria, Nsukka 410001, Nigeria
| |
Collapse
|
46
|
Leyderman M, Wilmore JR, Shope T, Cooney RN, Urao N. Impact of intestinal microenvironments in obesity and bariatric surgery on shaping macrophages. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00033. [PMID: 38037591 PMCID: PMC10683977 DOI: 10.1097/in9.0000000000000033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023]
Abstract
Obesity is associated with alterations in tissue composition, systemic cellular metabolism, and low-grade chronic inflammation. Macrophages are heterogenous innate immune cells ubiquitously localized throughout the body and are key components of tissue homeostasis, inflammation, wound healing, and various disease states. Macrophages are highly plastic and can switch their phenotypic polarization and change function in response to their local environments. Here, we discuss how obesity alters the intestinal microenvironment and potential key factors that can influence intestinal macrophages as well as macrophages in other organs, including adipose tissue and hematopoietic organs. As bariatric surgery can induce metabolic adaptation systemically, we discuss the potential mechanisms through which bariatric surgery reshapes macrophages in obesity.
Collapse
Affiliation(s)
- Michael Leyderman
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Joel R. Wilmore
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA
- Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Timothy Shope
- Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Robert N. Cooney
- Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY, USA
- Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Norifumi Urao
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, USA
- Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
47
|
Choi SI, Kim N, Nam RH, Jang JY, Kim EH, Ha S, Kang K, Lee W, Choi H, Kim YR, Seok YJ, Shin CM, Lee DH. The Protective Effect of Roseburia faecis Against Repeated Water Avoidance Stress-induced Irritable Bowel Syndrome in a Wister Rat Model. J Cancer Prev 2023; 28:93-105. [PMID: 37830115 PMCID: PMC10564633 DOI: 10.15430/jcp.2023.28.3.93] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/14/2023] Open
Abstract
Roseburia faecis, a butyrate-producing, gram-positive anaerobic bacterium, was evaluated for its usefulness against repeated water avoidance stress (WAS)-induced irritable bowel syndrome (IBS) in a rat model, and the underlying mechanism was explored. We divided the subjects into three groups: one without stress exposure, another subjected to daily 1-hour WAS for 10 days, and a third exposed to the same WAS regimen while also receiving two different R. faecis strains (BBH024 or R22-12-24) via oral gavage for the same 10-day duration. Fecal pellet output (FPO), a toluidine blue assay for mast cell infiltration, and fecal microbiota analyses were conducted using 16S rRNA metagenomic sequencing. Predictive functional profiling of microbial communities in metabolism was also conducted. FPO and colonic mucosal mast cell counts were significantly higher in the WAS group than in the control group (male, P = 0.004; female, P = 0.027). The administration of both BBH024 (male, P = 0.015; female, P = 0.022) and R22-12-24 (male, P = 0.003; female, P = 0.040) significantly reduced FPO. Submucosal mast cell infiltration in the colon showed a similar pattern in males. In case of fecal microbiota, the WAS with R. faecis group showed increased abundance of the Roseburia genus compared to WAS alone. Moreover, the expression of a gene encoding a D-methionine transport system substrate-binding protein was significantly elevated in the WAS with R. faecis group compared to that in the WAS (male, P = 0.028; female, P = 0.025) group. These results indicate that R. faecis is a useful probiotic for treating IBS and colonic microinflammation.
Collapse
Affiliation(s)
- Soo In Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Medical Device Development, Seoul National University College of Medicine, Seoul, Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jae Young Jang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Medical Device Development, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Hye Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - SungChan Ha
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | | | | | - HyeLim Choi
- Department of Biological Sciences and Institute of Microbiology, Seoul National University, Seoul, Korea
| | - Yeon-Ran Kim
- Department of Biological Sciences and Institute of Microbiology, Seoul National University, Seoul, Korea
| | - Yeong-Jae Seok
- Department of Biological Sciences and Institute of Microbiology, Seoul National University, Seoul, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
48
|
O’Reilly C, Mills S, Rea MC, Lavelle A, Ghosh S, Hill C, Ross RP. Interplay between inflammatory bowel disease therapeutics and the gut microbiome reveals opportunities for novel treatment approaches. MICROBIOME RESEARCH REPORTS 2023; 2:35. [PMID: 37849974 PMCID: PMC7615213 DOI: 10.20517/mrr.2023.41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/19/2023]
Abstract
Inflammatory bowel disease (IBD) is a complex heterogeneous disorder defined by recurring chronic inflammation of the gastrointestinal tract, attributed to a combination of factors including genetic susceptibility, altered immune response, a shift in microbial composition/microbial insults (infection/exposure), and environmental influences. Therapeutics generally used to treat IBD mainly focus on the immune response and include non-specific anti-inflammatory and immunosuppressive therapeutics and targeted therapeutics aimed at specific components of the immune system. Other therapies include exclusive enteral nutrition and emerging stem cell therapies. However, in recent years, scientists have begun to examine the interplay between these therapeutics and the gut microbiome, and we present this information here. Many of these therapeutics are associated with alterations to gut microbiome composition and functionality, often driving it toward a "healthier profile" and preclinical studies have revealed that such alterations can play an important role in therapeutic efficacy. The gut microbiome can also improve or hinder IBD therapeutic efficacy or generate undesirable metabolites. For certain IBD therapeutics, the microbiome composition, particularly before treatment, may serve as a biomarker of therapeutic efficacy. Utilising this information and manipulating the interactions between the gut microbiome and IBD therapeutics may enhance treatment outcomes in the future and bring about new opportunities for personalised, precision medicine.
Collapse
Affiliation(s)
- Catherine O’Reilly
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61C996, Ireland
- Microbiology Department, University College Cork, Co. Cork T12TP07, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
- Authors contributed equally
| | - Susan Mills
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
- Authors contributed equally
| | - Mary C. Rea
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61C996, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - Aonghus Lavelle
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - Subrata Ghosh
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - Colin Hill
- Microbiology Department, University College Cork, Co. Cork T12TP07, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - R. Paul Ross
- Microbiology Department, University College Cork, Co. Cork T12TP07, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| |
Collapse
|
49
|
Xiao L, Tang R, Wang J, Wan D, Yin Y, Xie L. Gut microbiota bridges the iron homeostasis and host health. SCIENCE CHINA. LIFE SCIENCES 2023; 66:1952-1975. [PMID: 37515687 DOI: 10.1007/s11427-022-2302-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/08/2023] [Indexed: 07/31/2023]
Abstract
The gut microbiota acts as a symbiotic microecosystem that plays an indispensable role in the regulation of a number of metabolic processes in the host by secreting secondary metabolites and impacting the physiology and pathophysiology of numerous organs and tissues through the circulatory system. This relationship, referred to as the "gut-X axis", is associated with the development and progression of disorders, including obesity, fatty liver and Parkinson's disease. Given its importance, the gut flora is a vital research area for the understanding and development of the novel therapeutic approaches for multiple disorders. Iron is a common but necessary element required by both mammals and bacteria. As a result, iron metabolism is closely intertwined with the gut microbiota. The host's iron homeostasis affects the composition of the gut microbiota and the interaction between host and gut microbiota through various mechanisms such as nutrient homeostasis, intestinal peaceability, gut immunity, and oxidative stress. Therefore, understanding the relationship between gut microbes and host iron metabolism is not only of enormous significance to host health but also may offer preventative and therapeutic approaches for a number of disorders that impact both parties. In this review, we delve into the connection between the dysregulation of iron metabolism and dysbiosis of gut microbiota, and how it contributes to the onset and progression of metabolic and chronic diseases.
Collapse
Affiliation(s)
- Lanling Xiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Rui Tang
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China
| | - Jie Wang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
- Department of Stomatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, China.
| |
Collapse
|
50
|
Dantas Machado AC, Ramos SF, Gauglitz JM, Fassler AM, Petras D, Aksenov AA, Kim UB, Lazarowicz M, Barnard Giustini A, Aryafar H, Vodkin I, Warren C, Dorrestein PC, Zarrinpar A, Zarrinpar A. Portosystemic shunt placement reveals blood signatures for the development of hepatic encephalopathy through mass spectrometry. Nat Commun 2023; 14:5303. [PMID: 37652904 PMCID: PMC10471626 DOI: 10.1038/s41467-023-40741-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 08/09/2023] [Indexed: 09/02/2023] Open
Abstract
Elective transjugular intrahepatic portosystemic shunt (TIPS) placement can worsen cognitive dysfunction in hepatic encephalopathy (HE) patients due to toxins, including possible microbial metabolites, entering the systemic circulation. We conducted untargeted metabolomics on a prospective cohort of 22 patients with cirrhosis undergoing elective TIPS placement and followed them up to one year post TIPS for HE development. Here we suggest that pre-existing intrahepatic shunting predicts HE severity post-TIPS. Bile acid levels decrease in the peripheral vein post-TIPS, and the abundances of three specific conjugated di- and tri-hydroxylated bile acids are inversely correlated with HE grade. Bilirubins and glycerophosphocholines undergo chemical modifications pre- to post-TIPS and based on HE grade. Our results suggest that TIPS-induced metabolome changes can impact HE development, and that pre-existing intrahepatic shunting could be used to predict HE severity post-TIPS.
Collapse
Affiliation(s)
| | - Stephany Flores Ramos
- Division of Gastroenterology and Hepatology, University of California, San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Julia M Gauglitz
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Anne-Marie Fassler
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Daniel Petras
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- CMFI Cluster of Excellence, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Alexander A Aksenov
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Chemistry, University of Connecticut, Storrs, CT, USA
| | - Un Bi Kim
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Michael Lazarowicz
- Department of Radiology, Division of Interventional Radiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Abbey Barnard Giustini
- Division of Gastroenterology and Hepatology, University of California, San Diego, La Jolla, CA, USA
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Puget Sounds Health Care System, Seattle, WA, USA
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Hamed Aryafar
- San Diego Imaging, San Diego, CA, USA
- Departments of Radiology, University of California San Diego Medical Center, La Jolla, CA, USA
| | - Irine Vodkin
- Division of Gastroenterology and Hepatology, University of California, San Diego, La Jolla, CA, USA
| | - Curtis Warren
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
- Center for Computational Mass Spectrometry, University of California, San Diego, La Jolla, CA, USA
| | - Ali Zarrinpar
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA.
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
| | - Amir Zarrinpar
- Division of Gastroenterology and Hepatology, University of California, San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA.
- Jennifer Moreno Department of Veterans Affairs Medical Center, La Jolla, CA, USA.
- Institute of Diabetes and Metabolic Health, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|