1
|
Xu S, Yue H, Zhang T, Zhou Z, Wang B, Ou Y, Deng S, Yin J, Zheng S. Parental high-fat diet induces upregulation of macrophage receptor with collagenous structure expression and exacerbates colorectal inflammation via the nuclear factor kappa-B pathway in offspring. J Nutr Biochem 2025; 142:109918. [PMID: 40254038 DOI: 10.1016/j.jnutbio.2025.109918] [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: 12/15/2024] [Revised: 03/20/2025] [Accepted: 04/04/2025] [Indexed: 04/22/2025]
Abstract
Parental high-fat diet (HFD) increases offspring's susceptibility to colorectal inflammation, but the underlying mechanism remains unclear. Using mouse models, we compared colorectal inflammation between offspring of HFD-fed and normal diet-fed parents. Histological analysis and immunostaining revealed that offspring of HFD-fed parents exhibited shortened colorectal length, decreased goblet cells, and reduced tight junction protein expression, particularly when maintained on HFD. RNA sequencing of colorectal tissue identified elevated expression of macrophage receptor with collagenous structure (MARCO) in these offspring. Immunofluorescence co-localization staining confirmed increased MARCO-positive macrophages in their colorectal tissue. Notably, switching offspring to normal diet partially alleviated these inflammatory responses, although some manifestations remained. Further investigation showed that high-lipid stimulation increased MARCO expression in macrophages and promoted inflammatory cytokine secretion through nuclear factor kappa-B (NF-κB) pathway activation. In vitro experiments demonstrated that MARCO knockdown inhibited the expression of inflammatory cytokines and prevented tight junction protein destruction in cocultured intestinal cells. Our findings reveal that parental HFD induces MARCO upregulation in offspring's colorectal macrophages and exacerbates colorectal inflammation through the NF-κB pathway, providing new insights into the mechanism by which parental HFD affects offspring's intestinal health.
Collapse
Affiliation(s)
- Shenghao Xu
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hui Yue
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ting Zhang
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhirui Zhou
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China; Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Bingbing Wang
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yinhua Ou
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Sanhua Deng
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jianbin Yin
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China; Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| | - Shimin Zheng
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Li X, Zhao Y, Liu M, Zhang M, Zhu Z, Liao A, Pan L, Lv X, Liu F, Huang J. Fermented Wheat Germ Ameliorates High-Fat Diet-Induced Maternal Obesity in Rats: Insights from Microbiome and Metabolomics. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12666-12678. [PMID: 40364743 DOI: 10.1021/acs.jafc.4c10847] [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: 05/15/2025]
Abstract
Maternal obesity significantly increases the risk of adverse outcomes for the mother and fetus. Fermented wheat germ (FWG) has demonstrated the potential to improve metabolic disorders, yet its effects have not been explored in maternal obesity models. This study investigated the ameliorating impact of FWG in rats with maternal obesity, focusing on its mechanisms through biochemical, gut microbiome, and serum metabolomics analysis. The results demonstrated that FWG was more effective than wheat germ in reducing body weight gain and fat accumulation, improving glycolipid metabolism disorders, and alleviating inflammation. Specifically, FWG modulated the composition of gut microbiota by fostering the growth of beneficial bacteria (e.g., Corynebacterium) while suppressing genera associated with maternal obesity (e.g., Blautia, Akkermansia, Dorea_A, and Faecousia). Furthermore, FWG modified high-fat diet-induced metabolites, primarily affecting pyrimidine metabolism and amino acid metabolism. These findings suggest that FWG may serve as a promising dietary intervention for mitigating maternal obesity and improving pregnancy outcomes.
Collapse
Affiliation(s)
- Xiaolin Li
- College of Grain and Food Science, Henan University of Technology, Zhengzhou 450001, China
| | - Yingyu Zhao
- College of Grain and Food Science, Henan University of Technology, Zhengzhou 450001, China
| | - Ming Liu
- College of Grain and Food Science, Henan University of Technology, Zhengzhou 450001, China
| | - Mingyi Zhang
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Zhitong Zhu
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Aimei Liao
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Long Pan
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Xin Lv
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Feng Liu
- Baolingbao Biology Co., Ltd., Dezhou, 253000, China
| | - Jihong Huang
- College of Grain and Food Science, Henan University of Technology, Zhengzhou 450001, China
| |
Collapse
|
3
|
Xu Z, Li L, Cheng L, Gu Z, Hong Y. Maternal obesity and offspring metabolism: revisiting dietary interventions. Food Funct 2025; 16:3751-3773. [PMID: 40289678 DOI: 10.1039/d4fo06233g] [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: 04/30/2025]
Abstract
Maternal obesity increases the risk of metabolic disorders in offspring. Understanding the mechanisms underlying the transgenerational transmission of metabolic diseases is important for the metabolic health of future generations. More research is needed to elucidate the mechanisms underlying the associated risks and their clinical implications because of the inherently complex nature of transgenerational metabolic disease transmission. Diet is a well-recognized risk factor for the development of obesity and other metabolic diseases, and rational dietary interventions are potential therapeutic strategies for their prevention. Despite extensive research on the physiological effects of diet on health and its associated mechanisms, little work has been devoted to understanding the effects of early-life dietary interventions on the metabolic health of offspring. In addition, existing dietary interventions are insufficient to meet clinical needs. Here, we discuss the literature on the effects of maternal obesity on the metabolic health of offspring, focusing on the mechanisms underlying the transgenerational transmission of metabolic diseases. We revisit current dietary interventions and describe their strengths and weaknesses in ameliorating maternal obesity-induced metabolism-related disorders in offspring. We also propose innovative strategies, such as the use of precision nutrition and fecal microbiota transplantation, which may limit the vicious cycle of intergenerational metabolic disease transmission.
Collapse
Affiliation(s)
- Zhiqiang Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Lingjin Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Jiaxing Institute of Future Food, Jiaxing 314050, China
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi, 214122, China
| | - Li Cheng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Jiaxing Institute of Future Food, Jiaxing 314050, China
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi, 214122, China
| | - Zhengbiao Gu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Jiaxing Institute of Future Food, Jiaxing 314050, China
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi, 214122, China
| | - Yan Hong
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Jiaxing Institute of Future Food, Jiaxing 314050, China
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi, 214122, China
| |
Collapse
|
4
|
Chen Y, Chen Z, Liang L, Li J, Meng L, Yuan W, Xie B, Zhang X, Feng L, Jia Y, Fu Z, Su P, Tong Z, Zhong J, Liu X. Multi-kingdom gut microbiota dysbiosis is associated with the development of pulmonary arterial hypertension. EBioMedicine 2025; 115:105686. [PMID: 40220715 PMCID: PMC12013117 DOI: 10.1016/j.ebiom.2025.105686] [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: 09/10/2024] [Revised: 03/07/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Gut microbiota dysbiosis has been implicated in pulmonary arterial hypertension (PAH). However, the exact roles and underlying mechanisms of multi-kingdom gut microbiota, including bacteria, archaea, and fungi, in PAH remain largely unclear. METHODS The shotgun metagenomics was used to analyse multi-kingdom gut microbial communities in patients with idiopathic PAH (IPAH) and healthy controls. Furthermore, fecal microbiota transplantation (FMT) was performed to transfer gut microbiota from IPAH patients or monocrotaline (MCT)-PAH rats to normal rats and from normal rats to MCT-PAH rats. FINDINGS Gut microbiota analysis revealed substantial alterations in the bacterial, archaeal, and fungal communities in patients with IPAH compared with healthy controls. Notably, FMT from IPAH patients or MCT-PAH rats induced PAH phenotypes in recipient rats. More intriguingly, FMT from normal rats to MCT-PAH rats significantly ameliorated PAH symptoms; restored gut bacteria, archaea, and fungi composition; and shifted the plasma metabolite profiles of MCT-PAH rats toward those of normal rats. In parallel, RNA-sequencing analysis demonstrated the expression of genes involved in key signalling pathways related to PAH. A panel of multi-kingdom markers exhibited superior diagnostic accuracy compared with single-kingdom panels for IPAH. INTERPRETATION Our findings established an association between multi-kingdom gut microbiota dysbiosis and PAH, thereby indicating the therapeutic potential of FMT in PAH. More importantly, apart from gut bacteria, gut archaea and fungi were also significantly associated with PAH pathogenesis, highlighting their indispensable role in PAH. FUNDING This work was supported by Noncommunicable Chronic Diseases-National Science and Technology Major Projects No. 2024ZD0531200, No. 2024ZD0531201 (Research on Prevention and Treatment of Cancer, Cardiovascular and Cerebrovascular Diseases, Respiratory Diseases, and Metabolic Diseases), the National Natural Science Foundation of China of China (No. 82170302, 82370432), Financial Budgeting Project of Beijing Institute of Respiratory Medicine (Ysbz2025004, Ysbz2025007), National clinical key speciality construction project Cardiovascular Surgery, Reform and Development Program of Beijing Institute of Respiratory Medicine (Ggyfz202417, Ggyfz202501), Clinical Research Incubation Program of Beijing Chaoyang Hospital Affiliated to Capital Medical University (CYFH202209).
Collapse
Affiliation(s)
- Yihang Chen
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zhenzhen Chen
- Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Lirong Liang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Clinical Epidemiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 100020, Beijing, China
| | - Jifeng Li
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Liukun Meng
- Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Wen Yuan
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Boqia Xie
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xun Zhang
- Department of Pharmacy, Chinese PLA General Hospital, 100039, Beijing, China
| | - Lin Feng
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Clinical Epidemiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, 100020, Beijing, China
| | - Yanxiong Jia
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zhou Fu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Pixiong Su
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zhaohui Tong
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Jiuchang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Xiaoyan Liu
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China; Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
5
|
Li X, Zhao Y, Huang J. Effects of fermented wheat germ on the placenta of high-fat diet-induced obese maternal rats: morphology, metabolism, and nutrient transport. Food Funct 2025; 16:2303-2315. [PMID: 39981968 DOI: 10.1039/d4fo05828c] [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: 02/22/2025]
Abstract
Maternal obesity impairs placental function, affecting fetal growth and long-term health. Although fermented wheat germ (FWG) provides health benefits, its impact on maternal obesity-related metabolic disorders and placental function remains unclear. This study investigated FWG's effects on placental morphology, metabolism, and nutrient transport in high-fat diet (HFD)-induced obese maternal rats. Wheat germ (WG) and FWG were administered from model induction, with a 45% HFD-fed for 10 weeks before conception and continued until gestational day 19.5. Results revealed that WG and FWG supplementation alleviated maternal metabolic abnormalities and mitigated placental structural damage. Additionally, this supplementation reduced placental lipid accumulation, oxidative stress, and inflammation while regulating nutrient transporter mRNA expression and inhibiting mTOR signaling activation. Compared with WG, FWG more effectively reduced maternal obesity and optimized placental nutrient transport. These findings suggest that FWG is a promising dietary intervention for disrupting the maternal obesity cycle and enhancing maternal-fetal health by alleviating obesity, mitigating metabolic dysfunction, and modulating placental morphology and function.
Collapse
Affiliation(s)
- Xiaolin Li
- College of Grain and Food Science, Henan University of Technology, Zhengzhou 450001, China.
- Food Laboratory of Zhongyuan, Henan University of Technology, Zhengzhou 450001, China
| | - Yingyu Zhao
- College of Grain and Food Science, Henan University of Technology, Zhengzhou 450001, China.
- Food Laboratory of Zhongyuan, Henan University of Technology, Zhengzhou 450001, China
| | - Jihong Huang
- College of Grain and Food Science, Henan University of Technology, Zhengzhou 450001, China.
- State Key Laboratory of Crop Stress Adaptation and Improvement, College of Agriculture, Henan University, Kaifeng 475004, China
- Food Laboratory of Zhongyuan, Henan University of Technology, Zhengzhou 450001, China
- School of Food and Pharmacy, Xuchang University, Xuchang 461000, China
| |
Collapse
|
6
|
Ge D, Zheng L, Liu L, Chen X, Zhou J, Ge H, Guo L, Hua H, Wang R, Zhang L. The dynamics impact of phlorizin on gut microbiota and metabolites in an in vitro fermentation model. Food Res Int 2025; 205:115930. [PMID: 40032457 DOI: 10.1016/j.foodres.2025.115930] [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: 11/14/2024] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025]
Abstract
The multiple beneficial effects, but low bioavailability of phlorizin (PHZ) have sparked discussion about its role in interaction with the gut microbiota. In this study, the effects of PHZ on the fecal microbiota animals of different origins were investigated using an in vitro fermentation model. In the fermentation system of PHZ using SD rat feces, the dynamic variations of the bacterial profile, SCFAs, and organic acids were detected using 16S rRNA gene sequencing, GC-MS, and LC-MS/MS. The results showed that PHZ treatment significantly increased the phylum Bacteroidota and transiently reduced Firmicutes at 6 h. At the genus level, PHZ consistently increased the abundance of Lactobacillus (especially Lactobacillus johnsonii), significantly decreased the abundance of Ligilactobacillus and Limosilactobacillus, and temporarily suppressed Streptococcus after 12 h. Similarly, in the fermentation system using db/db mouse feces, PHZ enriched the abundance of Lactobacillus and Lactobacillus johnsonii. Monoculture of Lactobacillus johnsonii ATCC 33200 showed that PHZ could directly stimulate its growth. Meanwhile, we found that PHZ could significantly increase the production of butyric, isobutyric, isovaleric, valeric, and caproic acids. Organic acid analysis showed an increasing trend in succinic acid and a significant reduction in L-malic acid in the post-PHZ group. Correlation analysis revealed that the abundance of Lactobacillus positively correlated with the concentration of SCFAs and succinic acid, while negatively correlated with L-malic acid. These findings suggest that PHZ may regulate intestinal balance by promoting Lactobacillus johnsonii growth and modulating SCFA and specific organic acid levels. Our study highlights that natural polyphenol PHZ has a health-promoting potential by modulating gut microbiota.
Collapse
Affiliation(s)
- Dingzuo Ge
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), China
| | - Luyao Zheng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Sichuan Institute for Translational Chinese Medicine, Chengdu, China
| | - Xin Chen
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiawei Zhou
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Han Ge
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liqiang Guo
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Hua
- Sichuan Institute for Translational Chinese Medicine, Chengdu, China; Sichuan Academy of Chinese Medical Sciences, Chengdu, China.
| | - Ruirui Wang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), China.
| | - Lei Zhang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine), China.
| |
Collapse
|
7
|
Xu H, Feng R, Ye M, Hu J, Lu J, Wang J, Zuo H, Zhao Y, Song J, Jiang J, Zhou Y, Wang Y. Multiple Enzymes Expressed by the Gut Microbiota Can Transform Typhaneoside and Are Associated with Improving Hyperlipidemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411770. [PMID: 39840606 PMCID: PMC11904961 DOI: 10.1002/advs.202411770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/10/2024] [Indexed: 01/23/2025]
Abstract
The mechanism of multiple enzymes mediated drug metabolism in gut microbiota is still unclear. This study explores multiple enzyme interaction process of typhactyloside (TYP) with gut microbiota and its lipid-lowering pharmacological activity. TYP, with bioavailability of only 2.78%, is an active component of Typha angustifolia L. and Pushen capsules which is clinically treated for hyperlipidemia. The metabolic process of TYP is identified, and key enzymes involved in TYP metabolism are validated through gene knockout and overexpression techniques. Through overexpressing α-rhamnosidase (Rha) in Escherichia coli, TYP is verified to metabolize into isorhamnetin-3-O-neohesperidin (M1) and isorhamnetin-3-O-glucoside (M2) after removing rhamnose through Rha. Besides, knockout of β-glucosidase (Glu) confirms that TYP generates M3 through Glu after removing glucose. Combined with molecular docking, M3 is transformed to generate 3,4-dihydroxyphenylacetic acid (M4), protocatechuic acid (M5), and 3-hydroxyphenylacetic acid (M6) through flavonoid reductase (Flr) and chalcone isomerase (Chi). In conclusion, multiple enzymes involved in TYP metabolism (Rha/Glu→Flr→Chi) are identified. Through in vivo experiments, combined use of M3 and M5 also shows excellent anti-hyperlipidemia efficacy. This is the first study on complex metabolism mechanism and pharmacological activity of natural flavonoids mediated by multiple enzymes, which provide insight to investigate analogous natural products.
Collapse
Affiliation(s)
- Hui Xu
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Ru Feng
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Meng‐Liang Ye
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Jia‐Chun Hu
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Jin‐Yue Lu
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Jing‐Yue Wang
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Heng‐Tong Zuo
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Yi Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Jian‐Ye Song
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Jian‐Dong Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Yun‐Zhi Zhou
- Emergency General HospitalNational Research Center for Emergency MedicineBeijing100028China
| | - Yan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia MedicaChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| |
Collapse
|
8
|
Yan F, Wang X, Du Y, Zhao Z, Shi L, Cao T, Shen Y, Sun L, Liu X. Pumpkin Soluble Dietary Fiber instead of Insoluble One Ameliorates Hyperglycemia via the Gut Microbiota-Gut-Liver Axis in db/db Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:1293-1307. [PMID: 39811930 DOI: 10.1021/acs.jafc.4c08986] [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: 01/16/2025]
Abstract
Pumpkin extract has been shown to alleviate hyperglycemic symptoms by improving glucose metabolism disorders. However, the specific active components responsible for its hypoglycemic effects and the underlying molecular mechanisms remain unclear. In this study, db/db mice underwent a 4-week dietary intervention with two pumpkin flours (PF1 and PF2), total dietary fiber (TDF), soluble dietary fiber (SDF), and insoluble dietary fiber (IDF), with acarbose serving as a positive control. Our results revealed that pumpkin components significantly altered the gut microbiota, characterized by a reduction in diabetes-related bacteria and an increase in short-chain fatty acid (SCFA)-producing bacteria, including Bacteroides, Akkermansia, and Lachnospiraceae_NK4A136 group. Additionally, pumpkin components significantly increased fecal SCFA levels and upregulated the expression of SCFA receptor GPR43, potentially promoting GLP-1 secretion. Notably, pumpkin components significantly reduced fasting blood glucose and serum insulin levels and inhibited gluconeogenesis. This effect may be ascribed to the inhibition of the cAMP/PKA/CREB signaling pathway coupled with the activation of the PI3K/AKT signaling pathway. Our research indicated that pumpkin flour and dietary fiber alleviated hyperglycemia through the gut-liver axis, with SDF contributing the most to the hypoglycemic effect. These findings suggest that pumpkin components may serve as an adjunct nutritional intervention to ameliorate hyperglycemia.
Collapse
Affiliation(s)
- Fanghua Yan
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Xinze Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Yue Du
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Zhongna Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Libing Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Tengzheng Cao
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Yajun Shen
- Yulin Keshangying Food Co., Ltd, Yulin, 719000 Shaanxi, China
| | - Lijun Sun
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| |
Collapse
|
9
|
Li B, Xiong Y, Guo D, Deng G, Wu H. The gut-reproductive axis: Bridging microbiota balances to reproductive health and fetal development. Int Immunopharmacol 2025; 144:113627. [PMID: 39579544 DOI: 10.1016/j.intimp.2024.113627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024]
Abstract
The gut microbiota is a highly complex microbial community residing in the digestive tract of humans and animals, closely linked to host health. Dysbiosis within the gut microbiota has been associated with various diseases. Moreover, it interacts with the female reproductive system's microbiota, influencing maternal reproductive homeostasis. Although the gut microbiota holds potential for treating reproductive system diseases and modulating offspring fertility, research in this domain remains limited. This review examines the relationship between both balanced and imbalanced gut microbiota and reproductive system diseases, as well as their effects on fetal development. It is highlighted that dysbiosis in the gut microbiota may contribute to several reproductive conditions, including polycystic ovary syndrome (PCOS), preeclampsia (PE), endometriosis, gestational diabetes, and reproductive cancers. The abundance of specific gut microbial species or interactions among various species can influence the reproductive system through hormonal pathways and other mechanisms, ultimately affecting pregnancy outcomes and fetal health. Therefore, the concept of the gut-reproductive axis is proposed, emphasizing the significant role of maternal gut microbiota in shaping fetal development, metabolic capacity, and immunity.
Collapse
Affiliation(s)
- Bohao Li
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Guohong Deng
- Department of Infectious Diseases, First Affiliated Hospital, Army Medical University, Chongqing, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing 401331, China; Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
10
|
Qin X, Zhang M, Chen S, Tang Y, Cui J, Ding G. Short-chain fatty acids in fetal development and metabolism. Trends Mol Med 2024:S1471-4914(24)00329-0. [PMID: 39694776 DOI: 10.1016/j.molmed.2024.11.014] [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: 08/31/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
Short-chain fatty acids (SCFAs), primarily derived from gut microbiota, play a role in regulating fetal development; however, the mechanism remains unclear. Fetal SCFAs levels depends on maternal SCFAs transported via the placenta. Metabolic stress, particularly from diabetes and obesity, can disrupt maternal SCFAs levels, impairing fetal metabolic reprogramming. Dysregulated SCFAs may negatively impact the development of the fetal cardiovascular, nervous, and immune systems, potentially contributing to adverse outcomes in adulthood. This review focuses on recent advances regarding the role of maternal SCFAs in shaping the metabolic profile of offspring, especially in the context of various maternal metabolic disorders. Given that SCFAs may influence fetal development through the placenta-embryo axis, targeted SCFAs supplementation could be a promising strategy against developmental diseases associated with intrauterine risk factors.
Collapse
Affiliation(s)
- Xueyun Qin
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Mo Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Shiting Chen
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Yunhui Tang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Jiajun Cui
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Guolian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
11
|
Tang M, Zhao L, Huang F, Wang T, Wu X, Chen S, Fu J, Jiang C, Wei S, Zeng X, Zhang X, Zhou X, Wei M, Li Z, Xiao G. Liuweizhiji Gegen-Sangshen beverage protects against alcoholic liver disease in mice through the gut microbiota mediated SCFAs/GPR43/GLP-1 pathway. Front Nutr 2024; 11:1495695. [PMID: 39734674 PMCID: PMC11673767 DOI: 10.3389/fnut.2024.1495695] [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: 09/13/2024] [Accepted: 11/27/2024] [Indexed: 12/31/2024] Open
Abstract
Introduction Alcoholic liver disease (ALD) is a pathological state of the liver caused by longterm alcohol consumption. Recent studies have shown that the modulation of the gut microbiota and its metabolic products, specifically the short-chain fatty acids (SCFAs), exert a critical role in the evolution and progression of ALD. The Liuweizhiji Gegen-Sangshen beverage (LGS), as a functional beverage in China, is derived from a traditional Chinese herbal formula and has been clinically applied for ALD treatment, demonstrating significant efficacy. However, the underlying mechanisms of LGS for alleviating ALD involving gut microbiota regulation remain unknown. Methods In this study, an ALD murine model based on the National Institute on Alcohol Abuse and Alcoholism (NIAAA) method was established. Results The results showed that oral LGS treatment dose-dependently alleviated alcoholinduced liver injury and inflammation in mice through decreasing levels of ALT, AST and proinflammatory cytokines (TNF-α, IL-6, IL-1β). LGS significantly improved liver steatosis, enhanced activities of alcohol metabolizing enzymes (ALDH and ADH), and reduced the CYP2E1 activity. Notably, regarding most detected indices, the effect of LGS (particularly at medium and high dose) was comparable to the positive drug MTDX. Moreover, LGS had a favorable effect on maintaining intestinal barrier function through reducing epithelial injury and increasing expression of occludin. 16S rRNA sequencing results showed that LGS remarkably modulated gut microbiota structure in ALD mice via recovering alcohol-induced microbial changes and specifically mediating enrichment of several bacterial genera (Alloprevotella, Monoglobus, Erysipelatoclostridium Parasutterella, Harryflintia and unclassified_c_Clostridia). Further study revealed that LGS increased production of SCFAs of hexanoic acid in cecum, promoted alcohol-mediated reduction of GRP43 expression in ileum, and increased serum GLP-1 level. Discussion Overall, LGS exerts a remarkable protective effect on ALD mice through the gut microbiota mediated specific hexanoic acid production and GPR43/GLP-1 pathway.
Collapse
Affiliation(s)
- Mingyun Tang
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Long Zhao
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Fuchun Huang
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Tiangang Wang
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xu Wu
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Shanshan Chen
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Juan Fu
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Chaoli Jiang
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Shulin Wei
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy and Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xuseng Zeng
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoling Zhang
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Zhou
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Mei Wei
- Department of Hepatobiliary Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhi Li
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Integrated Traditional Chinese and Western Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Guohui Xiao
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
12
|
Ren J, Zhou L, Li S, Zhang Q, Xiao X. The roles of the gut microbiota, metabolites, and epigenetics in the effects of maternal exercise on offspring metabolism. Am J Physiol Endocrinol Metab 2024; 327:E760-E772. [PMID: 39535269 DOI: 10.1152/ajpendo.00200.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/20/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Metabolic diseases, including obesity, dyslipidemia, and type 2 diabetes, have become severe challenges worldwide. The Developmental Origins of Health and Disease (DOHaD) hypothesis suggests that an adverse intrauterine environment can increase the risk of metabolic disorders in offspring. Studies have demonstrated that maternal exercise is an effective intervention for improving the offspring metabolic health. However, the pathways through which exercise works are unclear. It has been reported that the gut microbiota mediates the effect of maternal exercise on offspring metabolism, and epigenetic modifications have also been proposed to be important molecular mechanisms. Microbial metabolites can influence epigenetics by providing substrates for DNA or histone modifications, binding to G-protein-coupled receptors to affect downstream pathways, or regulating the activity of epigenetic modifying enzymes. This review aims to summarize the intergenerational effect of maternal exercise and proposes that gut microbiota-metabolites-epigenetic regulation is an important mechanism by which maternal exercise improves offspring metabolism, which may yield novel targets for the early prevention and intervention of metabolic diseases.
Collapse
Affiliation(s)
- Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Shunhua Li
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Chanda W, Jiang H, Liu SJ. The Ambiguous Correlation of Blautia with Obesity: A Systematic Review. Microorganisms 2024; 12:1768. [PMID: 39338443 PMCID: PMC11433710 DOI: 10.3390/microorganisms12091768] [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: 07/30/2024] [Revised: 08/13/2024] [Accepted: 08/17/2024] [Indexed: 09/30/2024] Open
Abstract
Obesity is a complex and multifactorial disease with global epidemic proportions, posing significant health and economic challenges. Whilst diet and lifestyle are well-established contributors to the pathogenesis, the gut microbiota's role in obesity development is increasingly recognized. Blautia, as one of the major intestinal bacteria of the Firmicutes phylum, is reported with both potential probiotic properties and causal factors for obesity in different studies, making its role controversial. To summarize the current understanding of the Blautia-obesity correlation and to evaluate the evidence from animal and clinical studies, we used "Blautia" AND "obesity" as keywords searching through PubMed and SpringerLink databases for research articles. After removing duplicates and inadequate articles using the exclusion criteria, we observed different results between studies supporting and opposing the beneficial role of Blautia in obesity at the genus level. Additionally, several studies showed probiotic effectiveness at the species level for Blautia coccoides, B. wexlerae, B. hansenii, B. producta, and B. luti. Therefore, the current evidence does not demonstrate Blautia's direct involvement as a pathogenic microbe in obesity development or progression, which informs future research and therapeutic strategies targeting the gut Blautia in obesity management.
Collapse
Affiliation(s)
- Warren Chanda
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
- Pathology and Microbiology Department, School of Medicine and Health Sciences, Mulungushi University, Livingstone P.O. Box 60009, Zambia
| | - He Jiang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
- State Key Laboratory of Microbial Resources, and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
14
|
Raczkowska E, Serek P. Health-Promoting Properties and the Use of Fruit Pomace in the Food Industry-A Review. Nutrients 2024; 16:2757. [PMID: 39203893 PMCID: PMC11357471 DOI: 10.3390/nu16162757] [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: 08/01/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 09/03/2024] Open
Abstract
Fruit pomace, a by-product of the fruit industry, includes the skins, seeds, and pulp most commonly left behind after juice extraction. It is produced in large quantities: apple residues alone generate approximately 4 million tons of waste annually, which is a serious problem for the processing industry but also creates opportunities for various applications. Due to, among other properties, their high content of dietary fiber and polyphenolic compounds, fruit residues are used to design food with functional features, improving the nutritional value and health-promoting, technological, and sensory properties of food products. This article presents the health-promoting (antioxidant, antidiabetic, anti-inflammatory, and antibacterial) properties of fruit pomace. Moreover, the possibilities of their use in the food industry are characterized, with particular emphasis on bread, sweet snack products, and extruded snacks. Attention is paid to the impact of waste products from the fruit industry on the nutritional value and technological and sensory characteristics of these products. Fruit pomace is a valuable by-product whose use in the food industry can provide a sustainable solution for waste management and contribute to the development of functional food products with targeted health-promoting properties.
Collapse
Affiliation(s)
- Ewa Raczkowska
- Department of Human Nutrition, Faculty of Biotechnology and Food Science, Wroclaw University of Environmental and Life Sciences, 37 Chelmonskiego Street, 51-630 Wroclaw, Poland;
| | | |
Collapse
|
15
|
Mostafavi Abdolmaleky H, Zhou JR. Gut Microbiota Dysbiosis, Oxidative Stress, Inflammation, and Epigenetic Alterations in Metabolic Diseases. Antioxidants (Basel) 2024; 13:985. [PMID: 39199231 PMCID: PMC11351922 DOI: 10.3390/antiox13080985] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/05/2024] [Accepted: 08/11/2024] [Indexed: 09/01/2024] Open
Abstract
Gut dysbiosis, resulting from an imbalance in the gut microbiome, can induce excessive production of reactive oxygen species (ROS), leading to inflammation, DNA damage, activation of the immune system, and epigenetic alterations of critical genes involved in the metabolic pathways. Gut dysbiosis-induced inflammation can also disrupt the gut barrier integrity and increase intestinal permeability, which allows gut-derived toxic products to enter the liver and systemic circulation, further triggering oxidative stress, inflammation, and epigenetic alterations associated with metabolic diseases. However, specific gut-derived metabolites, such as short-chain fatty acids (SCFAs), lactate, and vitamins, can modulate oxidative stress and the immune system through epigenetic mechanisms, thereby improving metabolic function. Gut microbiota and diet-induced metabolic diseases, such as obesity, insulin resistance, dyslipidemia, and hypertension, can transfer to the next generation, involving epigenetic mechanisms. In this review, we will introduce the key epigenetic alterations that, along with gut dysbiosis and ROS, are engaged in developing metabolic diseases. Finally, we will discuss potential therapeutic interventions such as dietary modifications, prebiotics, probiotics, postbiotics, and fecal microbiota transplantation, which may reduce oxidative stress and inflammation associated with metabolic syndrome by altering gut microbiota and epigenetic alterations. In summary, this review highlights the crucial role of gut microbiota dysbiosis, oxidative stress, and inflammation in the pathogenesis of metabolic diseases, with a particular focus on epigenetic alterations (including histone modifications, DNA methylomics, and RNA interference) and potential interventions that may prevent or improve metabolic diseases.
Collapse
Affiliation(s)
- Hamid Mostafavi Abdolmaleky
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|