1
|
Yuan Y, Xue T, Ren M, Liu Y, Song Z, Xiong Z, Qin F. Design and application of graphene oxide-based molecularly imprinted covalent organic framework for simultaneous and precise recognition of bile acid metabolites. Mikrochim Acta 2025; 192:271. [PMID: 40163176 DOI: 10.1007/s00604-025-07120-1] [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: 03/17/2025] [Indexed: 04/02/2025]
Abstract
In this study a bile acid (BA)-imprinted covalent organic framework (COF) was constructed via Schiff base reactions, which integrated the advantages of both inherent structural stability of COF and exceptional selectivity of molecular imprinting technology. Besides, it was found that the addition of graphene oxide (GO) effectively increased the dispersibility of nanoparticles, ultimately resulting in a GO-based molecularly imprinted COF (GO@MICOF). The GO@MICOF was identified with the characteristics of excellent mass transfer performance (20.09 mg g-1), specific surface area (152.35 m2 g-1), selectivity (IFs = 2.4), and regeneration ability (n ≥ 10). By coupling the GO@MICOF-based pretreatment method with ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) analysis, sensitive and accurate validation results (LOQs, 0.01-2.5 µmol L-1; extraction efficiency, 81.1-118.9%) were obtained. Notably, the application of the proposed pretreatment techniques to metabolomics analysis holds great significance for the precision of metabolomics results. Sixteen BA metabolites were successfully quantified in rat liver and fecal samples, and some potential biomarkers and related metabolic pathways associated with postmenopausal osteoporosis had been identified. Therefore, the integrated analysis strategy has significant advantages in purifying complex biological samples and has great application prospects in targeted metabolomics research.
Collapse
Affiliation(s)
- Yue Yuan
- School of Pharmacy, Shenyang Pharmaceutical University, New Tech Development Zone, No. 26 Huatuo Street, High &Liaoning Province, 117004, Benxi, China
| | - Tianyi Xue
- School of Pharmacy, Shenyang Pharmaceutical University, New Tech Development Zone, No. 26 Huatuo Street, High &Liaoning Province, 117004, Benxi, China
| | - Mengxin Ren
- School of Pharmacy, Shenyang Pharmaceutical University, New Tech Development Zone, No. 26 Huatuo Street, High &Liaoning Province, 117004, Benxi, China
| | - Yanzhu Liu
- School of Pharmacy, Shenyang Pharmaceutical University, New Tech Development Zone, No. 26 Huatuo Street, High &Liaoning Province, 117004, Benxi, China
| | - Zhexue Song
- School of Pharmacy, Shenyang Pharmaceutical University, New Tech Development Zone, No. 26 Huatuo Street, High &Liaoning Province, 117004, Benxi, China
| | - Zhili Xiong
- School of Pharmacy, Shenyang Pharmaceutical University, New Tech Development Zone, No. 26 Huatuo Street, High &Liaoning Province, 117004, Benxi, China.
| | - Feng Qin
- School of Pharmacy, Shenyang Pharmaceutical University, New Tech Development Zone, No. 26 Huatuo Street, High &Liaoning Province, 117004, Benxi, China.
| |
Collapse
|
2
|
Liu X, Fang W, Pang S, Song G, Wang Y, Qi W. Total dietary fiber of tartary buckwheat alleviates T2DM through the IRS-1/PI3K/AKT pathway and gut microbiota-bile acids-TGR5/FXR axis in db/db mice. Int J Biol Macromol 2025:142145. [PMID: 40101829 DOI: 10.1016/j.ijbiomac.2025.142145] [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: 10/19/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Buckwheat dietary fiber has good hypoglycemic activity, with its water-soluble parts and modification widely studied, however, the specific effects and mechanisms of action regarding buckwheat total dietary fiber (BDF) to improve T2DM need to be further explored. In this study, we investigated the improvement of diabetes-related symptoms in db/db mice by 10 % BDF and explored the possible mechanisms. The results showed that hyperglycemia, hyperlipemia, insulin resistance, elevated body weight, severe inflammatory response and impaired intestinal function were significantly improved after 8 weeks of BDF intervention. Moreover, BDF was able to ameliorate the distribution of gut microbiota, enhance the bacteria associated with T2DM, and improve glucose metabolism by altering the levels of the metabolites of gut microbiota, bile acids (BAs), and their receptors, TGR5 and FXR. In addition, BDF activated the hepatic IRS-1/PI3K/AKT pathway which can reduce hepatic gluconeogenesis and promoted glycogen synthesis. Taken together, our results suggest that BDF can alleviate T2DM symptoms by activating the IRS-1/PI3K/AKT pathway and the gut microbiota-BAs-TGR5/FXR axis. The results of this study deepen the exploration of the active ingredients of cereals and suggesting a scientific basis for the development of glucose-lowering functional foods and nutraceuticals.
Collapse
Affiliation(s)
- Xinguo Liu
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Wei Fang
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China.
| | - Shaojie Pang
- Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Chaoyang, Beijing 100015, China
| | - Ge Song
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Yong Wang
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Wentao Qi
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| |
Collapse
|
3
|
Dong H, Liu X, Song G, Peng W, Sun X, Fang W, Qi W. Imbalance of Bile Acids Metabolism Mediated by Gut Microbiota Contributed to Metabolic Disorders in Diabetic Model Mice. BIOLOGY 2025; 14:291. [PMID: 40136546 PMCID: PMC11940414 DOI: 10.3390/biology14030291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025]
Abstract
Type 2 diabetes (T2D) is a chronic disease prevalent in the world, accompanied by a variety of diseases, endangering human health and safety. Bile acids (BAs) play an important role in the regulation of host glucose and lipid metabolism homeostasis, and are strictly regulated by gut microbiota. However, the relationship between key BAs, BAs transporters and signaling, as well as gut microbiota, and host metabolism in T2D remains elusive. In this study, 9-week-old db/db mice were used as diabetes model (db/db group, n = 10), and their wild-type (wt) littermates of same age were used as the healthy control (CON group, n = 10). After 8 weeks of feeding, the BA profiles and microbial composition in the colon, and gene expression level of BA regulatory factors were analyzed in the db/db and CON groups to explore the underlying mechanisms of T2D. Compared with healthy mice, the body weight, blood glucose and lipid levels of db/db mice were significantly increased. The concentrations of total BAs, primary BAs, conjugated BAs and non-12α-hydroxylated BAs (non-12-OH BAs) were significantly decreased, while Deoxycholic acid (DCA) in secondary BAs was increased in db/db group. Compared with wt mice, the synthesis of BAs in the liver was transformed from the alternative pathway to the classical pathway, and hepatic BAs transporters (NTCP, BSEP, MRP2, OATP-1 and OSTβ) and receptors (FXR and TGR5) were significantly down-regulated in the db/db mice. In the colon, the mRNA level of FXR was up-regulated, while TGR5 was down-regulated. The diabetic (db/db) mice presented a changed gut microbiota composition, including an increased abundance of secondary BAs-producing bacteria, Escherichia-Shigella, and a decreased the abundance of Akkermansia, which are involved in the synthesis of non-12-OH BAs. We further found that the reduced BA types in db/db mice were negatively correlated with metabolic-disorder-related indicators, while an increased DCA level had the opposite correlation. Our results shed light into how the imbalance of BAs' metabolism mediated by intestinal flora may be potential mechanisms of T2D.
Collapse
Affiliation(s)
- Hongwang Dong
- Key Laboratory of Grain and Oil Biotechnology, Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; (H.D.); (X.L.); (G.S.); (W.P.); (X.S.)
- College of Food Science and Engineering, Central South University of Forestry and Technology, National Engineering Research Center for Rice and Byproduct Deep Processing, Changsha 410004, China
| | - Xinguo Liu
- Key Laboratory of Grain and Oil Biotechnology, Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; (H.D.); (X.L.); (G.S.); (W.P.); (X.S.)
| | - Ge Song
- Key Laboratory of Grain and Oil Biotechnology, Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; (H.D.); (X.L.); (G.S.); (W.P.); (X.S.)
| | - Wenting Peng
- Key Laboratory of Grain and Oil Biotechnology, Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; (H.D.); (X.L.); (G.S.); (W.P.); (X.S.)
| | - Xihan Sun
- Key Laboratory of Grain and Oil Biotechnology, Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; (H.D.); (X.L.); (G.S.); (W.P.); (X.S.)
| | - Wei Fang
- Key Laboratory of Grain and Oil Biotechnology, Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; (H.D.); (X.L.); (G.S.); (W.P.); (X.S.)
| | - Wentao Qi
- Key Laboratory of Grain and Oil Biotechnology, Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; (H.D.); (X.L.); (G.S.); (W.P.); (X.S.)
- College of Food Science and Engineering, Central South University of Forestry and Technology, National Engineering Research Center for Rice and Byproduct Deep Processing, Changsha 410004, China
| |
Collapse
|
4
|
Wang Y, Yu J, Chen B, Jin W, Wang M, Chen X, Jian M, Sun L, Piao C. Bile acids as a key target: traditional Chinese medicine for precision management of insulin resistance in type 2 diabetes mellitus through the gut microbiota-bile acids axis. Front Endocrinol (Lausanne) 2024; 15:1481270. [PMID: 39720247 PMCID: PMC11666381 DOI: 10.3389/fendo.2024.1481270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease caused by insulin resistance (IR) and insufficient insulin secretion. Its characteristic pathophysiological processes involve the interaction of multiple mechanisms. In recent years, globally, the prevalence of T2DM has shown a sharp rise due to profound changes in socio-economic structure, the persistent influence of environmental factors, and the complex role of genetic background. It is worth noting that most T2DM patients show significant IR, which further exacerbates the difficulty of disease progression and prevention. In the process of extensively exploring the pathogenesis of T2DM, the dynamic equilibrium of gut microbes and its diverse metabolic activities have increasingly emphasized its central role in the pathophysiological process of T2DM. Bile acids (BAs) metabolism, as a crucial link between gut microbes and the development of T2DM, not only precisely regulates lipid absorption and metabolism but also profoundly influences glucose homeostasis and energy balance through intricate signaling pathways, thus playing a pivotal role in IR progression in T2DM. This review aims to delve into the specific mechanism through which BAs contribute to the development of IR in T2DM, especially emphasizing how gut microbes mediate the metabolic transformation of BAs based on current traditional Chinese medicine research. Ultimately, it seeks to offer new insights into the prevention and treatment of T2DM. Diet, genetics, and the environment intricately sculpt the gut microbiota and BAs metabolism, influencing T2DM-IR. The research has illuminated the significant impact of single herbal medicine, TCM formulae, and external therapeutic methods such as electroacupuncture on the BAs pool through perturbations in gut microbiota structure. This interaction affects glucose and lipid metabolism as well as insulin sensitivity. Additionally, multiple pathways including BA-FXR-SHP, BA-FXR-FGFR15/19, BA-FXR-NLRP3, BA-TGR5-GLP-1, BAs-TGR5/FXR signaling pathways have been identified through which the BAs pool significantly alter blood glucose levels and improve IR. These findings offer novel approaches for enhancing IR and managing metabolic disorders among patients with T2DM.
Collapse
Affiliation(s)
- Yu Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jing Yu
- Department of Endocrinology, the Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Binqin Chen
- Applicants with Equivalent Academic Qualifications for Master Degree, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Shenzhen Hospital (Futian), Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Wenqi Jin
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Meili Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xuenan Chen
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Mengqiong Jian
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
- Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Chunli Piao
- Shenzhen Hospital (Futian), Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
5
|
Donati Zeppa S, Gervasi M, Bartolacci A, Ferrini F, Patti A, Sestili P, Stocchi V, Agostini D. Targeting the Gut Microbiota for Prevention and Management of Type 2 Diabetes. Nutrients 2024; 16:3951. [PMID: 39599740 PMCID: PMC11597803 DOI: 10.3390/nu16223951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024] Open
Abstract
Type 2 diabetes (T2D) is a chronic metabolic disorder with a heterogeneous etiology encompassing societal and behavioral risk factors in addition to genetic and environmental susceptibility. The cardiovascular consequences of diabetes account for more than two-thirds of mortality among people with T2D. Not only does T2D shorten life expectancy, but it also lowers quality of life and is associated with extremely high health expenditures since diabetic complications raise both direct and indirect healthcare costs. An increasing body of research indicates a connection between T2D and gut microbial traits, as numerous alterations in the intestinal microorganisms have been noted in pre-diabetic and diabetic individuals. These include pro-inflammatory bacterial patterns, increased intestinal permeability, endotoxemia, and hyperglycemia-favoring conditions, such as the alteration of glucagon-like peptide-1 (GLP-1) secretion. Restoring microbial homeostasis can be very beneficial for preventing and co-treating T2D and improving antidiabetic therapy outcomes. This review summarizes the characteristics of a "diabetic" microbiota and the metabolites produced by microbial species that can worsen or ameliorate T2D risk and progression, suggesting gut microbiota-targeted strategies to restore eubiosis and regulate blood glucose. Nutritional supplementation, diet, and physical exercise are known to play important roles in T2D, and here their effects on the gut microbiota are discussed, suggesting non-pharmacological approaches that can greatly help in diabetes management and highlighting the importance of tailoring treatments to individual needs.
Collapse
Affiliation(s)
- Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
- Department of Human Science for Promotion of Quality of Life, University San Raffaele, 00166 Rome, Italy;
| | - Marco Gervasi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
| | - Alessia Bartolacci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
| | - Fabio Ferrini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
| | - Antonino Patti
- Sport and Exercise Sciences Research Unit, Department of Psychology, Educational Science and Human Movement, University of Palermo, 90144 Palermo, Italy;
| | - Piero Sestili
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
| | - Vilberto Stocchi
- Department of Human Science for Promotion of Quality of Life, University San Raffaele, 00166 Rome, Italy;
| | - Deborah Agostini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
| |
Collapse
|
6
|
Liu X, Xia N, Yu Q, Jin M, Wang Z, Fan X, Zhao W, Li A, Jiang Z, Zhang L. Silybin Meglumine Mitigates CCl 4-Induced Liver Fibrosis and Bile Acid Metabolism Alterations. Metabolites 2024; 14:556. [PMID: 39452937 PMCID: PMC11509150 DOI: 10.3390/metabo14100556] [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: 09/18/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Altered patterns of bile acids (BAs) are frequently present in liver fibrosis, and BAs function as signaling molecules to initiate inflammatory responses. Silybin meglumine (SLB-M) is widely used in treating various liver diseases including liver fibrosis. However, research on its effects on bile acid (BA) metabolism is limited. This study investigated the therapeutic effects of SLB-M on liver fibrosis and BA metabolism in a CCl4-induced murine model. METHODS A murine liver fibrosis model was induced by CCl4. Fibrosis was evaluated using HE, picrosirius red, and Masson's trichrome staining. Liver function was assessed by serum and hepatic biochemical markers. Bile acid (BA) metabolism was analyzed using LC-MS/MS. Bioinformatics analyses, including PPI network, GO, and KEGG pathway analyses, were employed to explore molecular mechanisms. Gene expression alterations in liver tissue were examined via qRT-PCR. RESULTS SLB-M treatment resulted in significant histological improvements in liver tissue, reducing collagen deposition and restoring liver architecture. Biochemically, SLB-M not only normalized serum liver enzyme levels (ALT, AST, TBA, and GGT) but also mitigated disruptions in both systemic and hepatic BA metabolism by increased unconjugated BAs like cholic acid and chenodeoxycholic acid but decreased conjugated BAs including taurocholic acid and taurodeoxycholic acid, compared to that in CCl4-induced murine model. Notably, SLB-M efficiently improved the imbalance of BA homeostasis in liver caused by CCl4 via activating Farnesoid X receptor. CONCLUSIONS These findings underscore SLB-M decreased inflammatory response, reconstructed BA homeostasis possibly by regulating key pathways, and gene expressions in BA metabolism.
Collapse
Affiliation(s)
- Xiaoxin Liu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Ninglin Xia
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Qinwei Yu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Ming Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Zifan Wang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Xue Fan
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Wen Zhao
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Anqin Li
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Zhenzhou Jiang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Luyong Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
7
|
Lai C, Chen L, Zhong X, Tian X, Zhang B, Li H, Zhang G, Wang L, Sun Y, Guo L. Long-term arsenic exposure decreases mice body weight and liver lipid droplets. ENVIRONMENT INTERNATIONAL 2024; 192:109025. [PMID: 39317010 DOI: 10.1016/j.envint.2024.109025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/09/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
Arsenic (As) is a widespread global pollutant, and there is significant controversy surrounding its complex relationship with obesity, primarily focused on short-term exposure. Recognizing the prolonged nature of dietary arsenic exposure, this study involved feeding mice with arsenic-contained food for 14 months. The results showed that mice exposed to arsenic developed a non-alcoholic fatty liver condition, characterized by a light-yellow hue on the liver surface and various pathological alterations in the liver cells, including enlarged nuclei, cellular necrosis, inflammatory infiltration, dysfunctional mitochondria, and endoplasmic reticulum disorganization. There were also disruptions in biochemistry indices, with a significant increase in total cholesterol (TC) level and a decrease in high-density lipoprotein (HDL) level. However, some contradictory observations occurred, such as a significant decrease in body weight, triglyceride (TG) level, and the numbers of lipid droplets. Several genes related to lipid metabolism were tested, and a model was used to explain these discrepancies. Besides, examinations of the colon revealed compromised intestinal barrier function and signs of inflammation. Fecal 16S rRNA sequencing and pseudo-targeted metabolomics revealed disruptions in internal homeostasis, such as modules, nodes, connections, and lipid-related KEGG pathways. Fecal targeted metabolomics analyses of short-chain fatty acids (SCFAs) and bile acids (BAs) demonstrated a significant upregulation in three primary bile acids (CA, CDCA, TCDCA), four secondary bile acids (TUDCA, DCA, LCA, GUDCA), and total SCFAs level. Oxidative stress and inflammation were also evident. Additionally, based on correlation analysis and mediation analysis, it was assumed that changes in the microbiota (e.g., Dubosiella) can impact the liver metabolites (e.g., TGs) through alterations in fecal metabolites (e.g., LPCs). These findings provide a theoretical reference for the long-term effect of arsenic exposure on liver lipid metabolism.
Collapse
Affiliation(s)
- Chengze Lai
- Dongguan Key Laboratory of Public Health Laboratory Science, School of Public Health, Guangdong Medical University, Dongguan 523808, China; The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Linkang Chen
- Dongguan Key Laboratory of Public Health Laboratory Science, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Xiaoting Zhong
- Dongguan Key Laboratory of Public Health Laboratory Science, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Xianbing Tian
- School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Bin Zhang
- Dongguan Key Laboratory of Public Health Laboratory Science, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Hao Li
- Dongguan Key Laboratory of Public Health Laboratory Science, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Guiwei Zhang
- Shenzhen Academy of Metrology and Quality Inspection, Shenzhen 518000.China
| | - Liping Wang
- School of Nursing, Guangdong Medical University, Dongguan 523808, China
| | - Yanqin Sun
- Department of Pathology, School of Basic Medical Sciences, Guangdong Medical University, Dongguan 523808, China.
| | - Lianxian Guo
- Dongguan Key Laboratory of Public Health Laboratory Science, School of Public Health, Guangdong Medical University, Dongguan 523808, China; The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
8
|
Katsuyama H, Horinaka S, Hakoshima M, Adachi H, Yanai H. Retrospective Longitudinal Observational Study on the Long-Term Effects of Sodium-Glucose Cotransporter-2 Inhibitors on the Development of Metabolic Dysfunction-Associated Steatotic Liver Disease in Type 2 Diabetic Japanese Patients. J Clin Med 2024; 13:4929. [PMID: 39201071 PMCID: PMC11355713 DOI: 10.3390/jcm13164929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/02/2024] Open
Abstract
Background/Objectives: The health burden of metabolic dysfunction-associated fatty liver disease (MASLD) has been increasing lately. Cardiovascular disease (CVD) is the main cause of death in MASLD patients; therefore, the treatments for MASLD should improve both CV risk factors such as obesity, diabetes, and dyslipidemia, in addition to an improvement in liver function. The evidence on the long-term effects of sodium-glucose cotransporter 2 inhibitors (SGLT2is) on the progression of MASLD in Asian populations is very limited. Methods: The retrospective longitudinal study was performed by using the medical records at our institute. We picked up patients with type 2 diabetes who had taken SGLT2is for at least 3 years or longer between 1 April 2014 and 31 March 2018. We collected the data on metabolic parameters, including laboratory data and anthropometric parameters, and compared the data before and after the initiation of SGLT2is treatment. Results: During the observation period, 324 patients had taken SGLT2is for 3 years. Three-year SGLT2is treatment significantly reduced body weight, hemoglobin A1c, low-density lipoprotein cholesterol, triglyceride, and non-high-density lipoprotein cholesterol (non-HDL-C). Such favorable changes in serum lipids were remarkable in patients with statins. Furthermore, this treatment significantly improved liver function and the markers for hepatic steatosis and hepatic fibrosis. Conclusions: Considering that the development of CVD determines the prognosis of MASLD patients, long-term SGLT2is treatment may be an ideal therapy for MASLD patients.
Collapse
Affiliation(s)
| | | | | | | | - Hidekatsu Yanai
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Ichikawa 272-8516, Chiba, Japan; (H.K.); (S.H.); (M.H.); (H.A.)
| |
Collapse
|
9
|
Keivanlou MH, Amini-Salehi E, Sattari N, Hashemi M, Saberian P, Prabhu SV, Javid M, Mirdamadi A, Heidarzad F, Bakhshi A, Letafatkar N, Zare R, Hassanipour S, Nayak SS. Gut microbiota interventions in type 2 diabetes mellitus: An umbrella review of glycemic indices. Diabetes Metab Syndr 2024; 18:103110. [PMID: 39213690 DOI: 10.1016/j.dsx.2024.103110] [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: 11/21/2023] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND We aimed to explore how probiotics, prebiotics, or synbiotics impact glycemic indices in patients with diabetes mellitus. METHOD A comprehensive search was conducted on PubMed, Scopus, and Web of Science from inception up to April 2023. The random-effects model was employed for the study analysis. Furthermore, sensitivity and subgroup analyses were conducted to investigate potential sources of heterogeneity. AMSTAR2 checklist was used to determine the quality of studies. Comprehensive meta-analysis version 3 was used for the study analysis. RESULT A total of 31 studies were included in the final analysis. Based on the results of the meta-analysis, gut microbial therapy could significantly decrease serum fasting blood glucose levels in patients with type 2 diabetes mellitus (effect size: -0.211; 95 % CI: -0.257, -0.164; P < 0.001). Additionally, significant associations were also found between gut microbial therapy and improved serum levels of fasting insulin, glycated hemoglobin, and homeostatic model assessment for insulin resistance (effect size: -0.087; 95 % confidence interval: -0.120, -0.053; P < 0.001; effect size: -0.166; 95 % confidence interval: -0.200, -0.132; P < 0.001; effect size: -0.230; 95 % confidence interval: -0.288, -0.172; P < 0.001, respectively). CONCLUSION Our results revealed promising effects of gut microbiota modulation on glycemic profile of patients with type 2 diabetes mellitus. The use of these agents as additional treatments can be considered.
Collapse
Affiliation(s)
- Mohammad-Hossein Keivanlou
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Nazila Sattari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Hashemi
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Parsa Saberian
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Mona Javid
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Arian Mirdamadi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Forough Heidarzad
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Arash Bakhshi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Negin Letafatkar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Reza Zare
- Student Research Committee, Larestan University of Medical Sciences, Larestan, Iran
| | - Soheil Hassanipour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Sandeep Samethadka Nayak
- Department of Internal Medicine, Yale New Haven Health Bridgeport Hospital , Bridgeport, CT, USA
| |
Collapse
|
10
|
Vallianou NG, Kounatidis D, Psallida S, Vythoulkas-Biotis N, Adamou A, Zachariadou T, Kargioti S, Karampela I, Dalamaga M. NAFLD/MASLD and the Gut-Liver Axis: From Pathogenesis to Treatment Options. Metabolites 2024; 14:366. [PMID: 39057689 PMCID: PMC11278747 DOI: 10.3390/metabo14070366] [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: 05/14/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) poses an emerging threat topublic health. Nonalcoholic steatohepatitis (NASH) is reported to be the most rapidly rising cause of hepatocellular carcinoma in the western world. Recently, a new term has been proposed: metabolic dysfunction-associated steatotic liver disease (MASLD). The introduction of this new terminology has sparked a debate about the interchangeability of these terms. The pathogenesis of NAFLD/MASLD is thought to be multifactorial, involving both genetic and environmental factors. Among these factors, alterations in gut microbiota and gut dysbiosis have recently garnered significant attention. In this context, this review will further discuss the gut-liver axis, which refers to the bidirectional interaction between the human gut microbiota and the liver. Additionally, the therapeutic potential of probiotics, particularly next-generation probiotics and genetically engineered bacteria, will be explored. Moreover, the role of prebiotics, synbiotics, postbiotics, and phages as well as fecal microbiota transplantation will be analyzed. Particularly for lean patients with NAFLD/MASLD, who have limited treatment options, approaches that modify the diversity and composition of the gut microbiota may hold promise. However, due to ongoing safety concerns with approaches that modulate gut microbiota, further large-scale studies are necessary to better assess their efficacy and safety in treating NAFLD/MASLD.
Collapse
Affiliation(s)
- Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, Sismanogliou 1 Str., 15126 Athens, Greece
| | - Dimitris Kounatidis
- Department of Internal Medicine, Hippokration General Hospital, 114 Vassilissis Sofias Str., 11527 Athens, Greece;
| | - Sotiria Psallida
- Department of Microbiology, “KAT” General Hospital of Attica, 14561 Athens, Greece;
| | - Nikolaos Vythoulkas-Biotis
- First Department of Internal Medicine, Sismanogleio General Hospital, Sismanogliou 1 Str., 15126 Athens, Greece
| | - Andreas Adamou
- First Department of Internal Medicine, Sismanogleio General Hospital, Sismanogliou 1 Str., 15126 Athens, Greece
| | - Tatiana Zachariadou
- First Department of Internal Medicine, Sismanogleio General Hospital, Sismanogliou 1 Str., 15126 Athens, Greece
| | - Sofia Kargioti
- First Department of Internal Medicine, Sismanogleio General Hospital, Sismanogliou 1 Str., 15126 Athens, Greece
| | - Irene Karampela
- Second Department of Critical Care, Attikon University Hospital, 1 Rimini Str., 12462 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| |
Collapse
|
11
|
Nanaware PP, Khan ZN, Clement CC, Shetty M, Mota I, Seltzer ES, Dzieciatkowska M, Gamboni F, D'Alessandro A, Ng C, Nagayama M, Lichti CF, Soni RK, Jacob B Geri, Matei I, Lyden D, Longman R, Lu TT, Wan X, Unanue ER, Stern LJ, Santambrogio L. Role of the afferent lymph as an immunological conduit to analyze tissue antigenic and inflammatory load. Cell Rep 2024; 43:114311. [PMID: 38848214 PMCID: PMC11233987 DOI: 10.1016/j.celrep.2024.114311] [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: 10/16/2023] [Revised: 04/03/2024] [Accepted: 05/16/2024] [Indexed: 06/09/2024] Open
Abstract
The lymphatic fluid is the conduit by which part of the tissue "omics" is transported to the draining lymph node for immunosurveillance. Following cannulation of the pre-nodal cervical and mesenteric afferent lymphatics, herein we investigate the lymph proteomic composition, uncovering that its composition varies according to the tissue of origin. Tissue specificity is also reflected in the dendritic cell-major histocompatibility complex class II-eluted immunopeptidome harvested from the cervical and mesenteric nodes. Following inflammatory disruption of the gut barrier, the lymph antigenic and inflammatory loads are analyzed in both mice and subjects with inflammatory bowel diseases. Gastrointestinal tissue damage reflects the lymph inflammatory and damage-associated molecular pattern signatures, microbiome-derived by-products, and immunomodulatory molecules, including metabolites of the gut-brain axis, mapped in the afferent mesenteric lymph. Our data point to the relevance of the lymphatic fluid to probe the tissue-specific antigenic and inflammatory load transported to the draining lymph node for immunosurveillance.
Collapse
Affiliation(s)
- Padma P Nanaware
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Zohaib N Khan
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Cristina C Clement
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Madhur Shetty
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ines Mota
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ethan S Seltzer
- Pediatric Rheumatology and Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, New York NY 100021, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Fabia Gamboni
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Charles Ng
- Department of Pathology and Laboratory Medicine, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY 10065, USA
| | - Manabu Nagayama
- Division of Gastroenterology and Hepatology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY 10065, USA
| | - Cheryl F Lichti
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Rajesh K Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York 10032, NY, USA
| | - Jacob B Geri
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| | - David Lyden
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| | - Randy Longman
- Division of Gastroenterology and Hepatology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY 10065, USA
| | - Theresa T Lu
- Pediatric Rheumatology and Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, New York NY 100021, USA
| | - Xiaoxiao Wan
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Lawrence J Stern
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY 10065, USA.
| |
Collapse
|
12
|
Firrman J, Friedman ES, Hecht A, Strange WC, Narrowe AB, Mahalak K, Wu GD, Liu L. Preservation of conjugated primary bile acids by oxygenation of the small intestinal microbiota in vitro. mBio 2024; 15:e0094324. [PMID: 38727244 PMCID: PMC11237543 DOI: 10.1128/mbio.00943-24] [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: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 06/13/2024] Open
Abstract
Bile acids play a critical role in the emulsification of dietary lipids, a critical step in the primary function of the small intestine, which is the digestion and absorption of food. Primary bile acids delivered into the small intestine are conjugated to enhance functionality, in part, by increasing aqueous solubility and preventing passive diffusion of bile acids out of the gut lumen. Bile acid function can be disrupted by the gut microbiota via the deconjugation of primary bile acids by bile salt hydrolases (BSHs), leading to their conversion into secondary bile acids through the expression of bacterial bile acid-inducible genes, a process often observed in malabsorption due to small intestinal bacterial overgrowth. By modeling the small intestinal microbiota in vitro using human small intestinal ileostomy effluent as the inocula, we show here that the infusion of physiologically relevant levels of oxygen, normally found in the proximal small intestine, reduced deconjugation of primary bile acids, in part, through the expansion of bacterial taxa known to have a low abundance of BSHs. Further recapitulating the small intestinal bile acid composition of the small intestine, limited conversion of primary into secondary bile acids was observed. Remarkably, these effects were preserved among four separate communities, each inoculated with a different small intestinal microbiota, despite a high degree of taxonomic variability under both anoxic and aerobic conditions. In total, these results provide evidence for a previously unrecognized role that the oxygenated environment of the small intestine plays in the maintenance of normal digestive physiology. IMPORTANCE Conjugated primary bile acids are produced by the liver and exist at high concentrations in the proximal small intestine, where they are critical for proper digestion. Deconjugation of these bile acids with subsequent transformation via dehydroxylation into secondary bile acids is regulated by the colonic gut microbiota and reduces their digestive function. Using an in vitro platform modeling the small intestinal microbiota, we analyzed the ability of this community to transform primary bile acids and studied the effect of physiological levels of oxygen normally found in the proximal small intestine (5%) on this metabolic process. We found that oxygenation of the small intestinal microbiota inhibited the deconjugation of primary bile acids in vitro. These findings suggest that luminal oxygen levels normally found in the small intestine may maintain the optimal role of bile acids in the digestive process by regulating bile acid conversion by the gut microbiota.
Collapse
Affiliation(s)
- Jenni Firrman
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, US Department of Agriculture, Wyndmoor, Pennsylvania, USA
| | - Elliot S. Friedman
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Aaron Hecht
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William C. Strange
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adrienne B. Narrowe
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, US Department of Agriculture, Wyndmoor, Pennsylvania, USA
| | - Karley Mahalak
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, US Department of Agriculture, Wyndmoor, Pennsylvania, USA
| | - Gary D. Wu
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - LinShu Liu
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, US Department of Agriculture, Wyndmoor, Pennsylvania, USA
| |
Collapse
|
13
|
Nguyen Y, Rudd Zhong Manis J, Ronczkowski NM, Bui T, Oxenrider A, Jadeja RN, Thounaojam MC. Unveiling the gut-eye axis: how microbial metabolites influence ocular health and disease. Front Med (Lausanne) 2024; 11:1377186. [PMID: 38799150 PMCID: PMC11122920 DOI: 10.3389/fmed.2024.1377186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/19/2024] [Indexed: 05/29/2024] Open
Abstract
The intricate interplay between the gut microbiota and ocular health has surpassed conventional medical beliefs, fundamentally reshaping our understanding of organ interconnectivity. This review investigates into the intricate relationship between gut microbiota-derived metabolites and their consequential impact on ocular health and disease pathogenesis. By examining the role of specific metabolites, such as short-chain fatty acids (SCFAs) like butyrate and bile acids (BAs), herein we elucidate their significant contributions to ocular pathologies, thought-provoking the traditional belief of organ sterility, particularly in the field of ophthalmology. Highlighting the dynamic nature of the gut microbiota and its profound influence on ocular health, this review underlines the necessity of comprehending the complex workings of the gut-eye axis, an emerging field of science ready for further exploration and scrutiny. While acknowledging the therapeutic promise in manipulating the gut microbiome and its metabolites, the available literature advocates for a targeted, precise approach. Instead of broad interventions, it emphasizes the potential of exploiting specific microbiome-related metabolites as a focused strategy. This targeted approach compared to a precision tool rather than a broad-spectrum solution, aims to explore the therapeutic applications of microbiome-related metabolites in the context of various retinal diseases. By proposing a nuanced strategy targeted at specific microbial metabolites, this review suggests that addressing specific deficiencies or imbalances through microbiome-related metabolites might yield expedited and pronounced outcomes in systemic health, extending to the eye. This focused strategy holds the potential in bypassing the irregularity associated with manipulating microbes themselves, paving a more efficient pathway toward desired outcomes in optimizing gut health and its implications for retinal diseases.
Collapse
Affiliation(s)
- Yvonne Nguyen
- Mercer University School of Medicine, Macon, GA, United States
| | | | | | - Tommy Bui
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Allston Oxenrider
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Ravirajsinh N. Jadeja
- Biochemistry and Molecular Biology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Menaka C. Thounaojam
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| |
Collapse
|
14
|
Wang Z, Li J, Xu Y, Liu Y, Zhang Z, Xu Q, Lin J, Jiang Y, Wang Y, Jing J, Wang A, Meng X. Elevated gut microbiota metabolite bile acids confer protective effects on clinical prognosis in ischemic stroke patients. Front Neurosci 2024; 18:1388748. [PMID: 38650617 PMCID: PMC11033300 DOI: 10.3389/fnins.2024.1388748] [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: 02/20/2024] [Accepted: 03/28/2024] [Indexed: 04/25/2024] Open
Abstract
Background There is evidence of an association between the gut microbiota and progression of stroke. However, the relationship between gut microbial metabolites, specifically bile acids (BAs), and post-ischemic stroke disability and poor functional outcomes remains unexplored. Methods Patients with acute ischemic stroke (AIS) or transient ischemic attack (TIA) in the Third China National Stroke Registry were grouped according to total bile acid (TBA) quartile on admission. Association of TBA with disability and poor functional outcomes were evaluated using logistic regression models and restricted cubic splines. Results Data for 9,536 patients were included. After adjusting for confounders, the risks of disability and poor functional outcomes were significantly lower in the highest TBA quartile than in the lowest TBA quartile at the 3-month follow-up, with respective odds ratios (ORs) of 0.65 (95% confidence interval [CI] 0.55-0.78; p < 0.001) and 0.66 (95% CI 0.55-0.78, p < 0.001). Each standard deviation increase in the TBA level reduced the risks of disability and poor functioning outcomes by 10% (adjusted ORs 0.9 [95% CI 0.83-0.98; p = 0.01] and 0.9 [95% CI 0.83-0.97; p < 0.001], respectively). This association remained similar at the 1-year follow-up. After stratification by TOAST subtype, the risk of disability or a poor functional outcome in patients with the large-artery atherosclerosis or "other" subtype was significantly lower in the highest quartile than in the lowest quartile (p < 0.05). Conclusion Serum TBA is an independent risk factor for disability and poor functional outcomes after AIS or TIA, and exerts a protective effects on brain.
Collapse
Affiliation(s)
- Zhaobin Wang
- Affiliated Hospital of Hebei University, Baoding, China
- Clinical Medical College, Hebei University, Baoding, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Puyang Oilfield General Hospital, Puyang, China
| | - Jing Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yingxin Xu
- Department of Neurology, Beijing Daxing District People's Hospital, Beijing, China
| | - Ye Liu
- Department of Anaesthesiology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Zhe Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qin Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinxi Lin
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yong Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jing Jing
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Anxin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xia Meng
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Al-Dejeli AAB, Al-Mudhafar MA, Al-Sabri IKA. Adenosine triphosphate binding cassette transporters G5 and G8 early diagnostic tools for cardiovascular disease in human. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:262-267. [PMID: 38592987 DOI: 10.36740/wlek202402111] [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: 04/11/2024]
Abstract
OBJECTIVE Aim: The current study was designed to investigate the role of ABCG5 and ABCG5 in serum with normal and expected cardiac complaints with CVDs as individual early diagnostic tools. PATIENTS AND METHODS Materials and Methods: Data was collected in paper form and recorded from 100 healthy personals and 100 personals suspected with CVS after take the case history and clinical signs in private clinical hospital and the serum was collected for measurements the activity of ABCG5 and ABCG5 by used ELISA reader and the results illustrated that activity of ABCG5 and ABCG5 in all aged groups. RESULTS Results: Activity of ABCG5 and ABCG5 in all aged groups periods in patient person male and female significant decrease as compared with same age in same period of live, so that the researched depicted that can used the serum activity of ABCG5 and ABCG5 as a diagnostics tools for atherosclerotic cardiovascular disease. CONCLUSION Conclusions: We identified areas of further exploration on cholesterol transport related with CVD risk and concluded that changes in the Adenosine Triphosphate Binding Cassette transporters mainly G5 and G8 early diagnostic tools for cardiovascular disease in Human. We correlated areas of farther disquisition on nutrient cholesterol and CVD threat, in the included trials, healthy grown-ups consumed high doses of dietary cholesterol.
Collapse
|