1
|
Kordahi MC, Daniel N, Gewirtz AT, Chassaing B. Mucus-penetrating microbiota drive chronic low-grade intestinal inflammation and metabolic dysregulation. Gut Microbes 2025; 17:2455790. [PMID: 39865067 PMCID: PMC11776472 DOI: 10.1080/19490976.2025.2455790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 01/28/2025] Open
Abstract
Metabolic syndrome is, in humans, associated with alterations in the composition and localization of the intestinal microbiota, including encroachment of bacteria within the colon's inner mucus layer. Possible promoters of these events include dietary emulsifiers, such as carboxymethylcellulose (CMC) and polysorbate-80 (P80), which, in mice, result in altered microbiota composition, encroachment, low-grade inflammation and metabolic syndrome. While assessments of gut microbiota composition have largely focused on fecal/luminal samples, we hypothesize an outsized role for changes in mucus microbiota in driving low-grade inflammation and its consequences. In support of this notion, we herein report that both CMC and P80 led to stark changes in the mucus microbiome, markedly distinct from those observed in feces. Moreover, transfer of mucus microbiota from CMC- and P80-fed mice to germfree mice resulted in microbiota encroachment, low-grade inflammation, and various features of metabolic syndrome. Thus, we conclude that mucus-associated bacteria are pivotal determinants of intestinal inflammatory tone and host metabolism.
Collapse
Affiliation(s)
- Melissa C. Kordahi
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, CNRS UMR6047, Paris, France
- Mucosal microbiota in chronic inflammatory diseases, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris, France
| | - Noëmie Daniel
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, CNRS UMR6047, Paris, France
- Mucosal microbiota in chronic inflammatory diseases, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris, France
| | - Andrew T. Gewirtz
- Institute for Biomedical Sciences, Centre for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, CNRS UMR6047, Paris, France
- Mucosal microbiota in chronic inflammatory diseases, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris, France
- CHRU Nancy, IHU Infiny, Nancy, France
| |
Collapse
|
2
|
Luo F, Yang J, Song Z, Zhao Y, Wang P, Liu K, Mou X, Liu W, Li W. Renshen Zhuye decoction ameliorates high-fat diet-induced obesity and insulin resistance by modulating gut microbiota and metabolic homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156655. [PMID: 40120542 DOI: 10.1016/j.phymed.2025.156655] [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: 01/20/2025] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Obesity, characterized by excessive adipose tissue accumulation, has become a global health challenge with rapidly increasing prevalence. It contributes significantly to metabolic disorders including insulin resistance (IR). Renshen-zhuye decoction (RZD), a traditional Chinese medicine formula historically used for diabetes, shows potential for improving metabolic parameters, but its effects and mechanisms in obesity and insulin resistance remain unclear. PURPOSE This study aimed to evaluate the therapeutic benefits of RZD on obesity and insulin resistance, and to elucidate the underlying mechanisms through which it improves glucose and lipid metabolism. METHODS The role of RZD was evaluated in a high-fat diet (HFD) mouse model. The formula was characterized using UPLC-MS. Comprehensive analyses including histopathological staining, immunofluorescence, biochemical assays, 16S rRNA gene sequencing of gut microbiota, and non-targeted metabolomic analysis were performed. To validate the role of gut microbiota, we employed antibiotic treatment (ABX) to deplete intestinal flora and conducted fecal microbiota transplantation (FMT) experiments. RESULTS RZD treatment dose-dependently alleviated HFD-induced dyslipidemia and insulin resistance, improving glucose tolerance, insulin sensitivity, and energy expenditure. Gut microbiota analysis revealed that RZD significantly modulated the composition of intestinal flora and their metabolic profiles. Additionally, RZD reduced intestinal and systemic inflammation by enhancing intestinal barrier integrity, particularly through increased expression of tight junction proteins such as Occludin. Importantly, the beneficial effects of RZD on weight management and glucose homeostasis were antagonized by antibiotic intervention, while FMT experiments confirmed that these improvements were mediated through gut microbiota modulation. CONCLUSION This study provides new insights into RZD's modulatory effects on gut microbiota and subsequent improvements in obesity-related metabolic parameters. RZD alleviates HFD-induced obesity and insulin resistance in mice by modulating gut microbiota composition and function, which subsequently improves intestinal barrier integrity, reduces inflammation, and enhances metabolic homeostasis.
Collapse
Affiliation(s)
- Fei Luo
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Jie Yang
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Zhiping Song
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Yuan Zhao
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Panpan Wang
- Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou 310000, PR China
| | - Kaiyuan Liu
- Department of Endocrinology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310000, PR China
| | - Xin Mou
- Department of Endocrinology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310000, PR China.
| | - Wenhong Liu
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| | - Wei Li
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| |
Collapse
|
3
|
Zhang W, Yi C, Song Z, Yu B, Jiang X, Guo L, Huang S, Xia T, Huang F, Yan Y, Li H, Dai Y. Reshaping the gut microbiota: Tangliping decoction and its core blood-absorbed component quercetin improve diabetic cognitive impairment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156560. [PMID: 40058319 DOI: 10.1016/j.phymed.2025.156560] [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: 07/02/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is associated with an increased risk of cognitive decline, which can result in diabetic cognitive impairment (DCI). Recent studies have indicated that gut microbiota plays a significant role in the development of DCI. Tangliping Decoction (TLP), a traditional Chinese medicine compound, contains various active ingredients that have been shown to regulate the microecology of gut microbiota and potentially improve DCI. However, it remains unclear whether TLP can improve DCI by modulating gut microbiota, as well as which specific component is primarily responsible for these effects. PURPOSE Assess the impact of TLP on alleviating DCI and investigate the contribution of quercetin (QR), the core blood-absorbed component of TLP, in this process. and investigate the underlying mechanisms through which TLP and QR enhance DCI by modulating gut microbiota composition. STUDY DESIGN AND METHODS Initially, experiments such as morris water maze (MWM), morphological analysis, and 16S ribosomal RNA (16S rRNA) gene amplicon sequencing from DCI mice, were performed to validate the pharmacological efficacy of TLP in mitigating DCI. The results indicated that TLP possesses the capacity to modulate the composition and quantity of gut microbiota and safeguard the integrity of the gut barrier and brain barrier. Secondly, high performance liquid chromatography coupled with high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (HPLC-Q-TOF-MS/MS) combined with network pharmacology methods were used to screen for blood-absorbed components, suggesting that QR may be a potential core blood-absorbed component of TLP in the treatment of DCI. Subsequently, the pharmacological efficacy of QR in ameliorating DCI was confirmed, and the characteristics of gut microbiota as well as the permeability of the gut and brain barrier, were assessed. Finally, fecal microbiota transplantation (FMT) experiments were conducted, wherein fecal matter from TLP and QR-treated mice (donor mice) was transplanted into pseudo-sterile DCI mice with antibiotic-induced depletion of gut microbiota. This approach aimed to elucidate the specific mechanisms by which TLP and QR improve DCI through the modulation of the structure, composition, and abundance of gut microbiota. RESULTS TLP and QR have the potential to enhance learning and memory capabilities in DCI mice, as well as reduce homeostasis model assessment insulin resistance (HOMA-IR) and restore homeostasis model assessment-β function (HOMA- β), leading to increased fasting insulin (FIN) levels and decreased fasting blood glucose (FBG) levels. Simultaneously, the administration of FMT from donor mice to pseudo-sterile DCI mice has been shown to alter the composition and abundance of gut microbiota, leading to amelioration of pathological damage in the colon and hippocampal tissues. Ultimately, FMT utilizing fecal suspensions from donor mice treated with TLP and QR improved cognitive function in pseudo-sterile DCI mice, restore gut microbiota dysbiosis, and maintained the integrity of the gut and brain barriers. CONCLUSION The results of this study indicate that TLP and its core component, QR, which is absorbed into the bloodstream, improve DCI through a gut microbiota-dependent mechanism, providing further evidence for gut microbiota as a therapeutic target for DCI treatment.
Collapse
Affiliation(s)
- Wenlan Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chunmei Yi
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhijie Song
- Department of Rheumatology and Immunology, Chifeng Cancer Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, Chifeng, Inner Mongolia, China
| | - Bin Yu
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shanshan Huang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tong Xia
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fayin Huang
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yijing Yan
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huhu Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yongna Dai
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
4
|
Fu L, Baranova A, Cao H, Zhang F. Gut microbiome links obesity to type 2 diabetes: insights from Mendelian randomization. BMC Microbiol 2025; 25:253. [PMID: 40289103 PMCID: PMC12034155 DOI: 10.1186/s12866-025-03968-8] [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: 10/27/2024] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Research has established links between the gut microbiome (GM) and both obesity and type 2 diabetes (T2D), which is much discussed, but underexplored. This study employed body mass index (BMI) as the measurement of obesity to delve deeper into the correlations from a genetic perspective. METHODS We performed the Mendelian randomization (MR) analysis to examine the causal effects of GM on T2D and BMI, and vice versa. Genome-wide association study (GWAS) summary datasets were utilized for the analysis, including T2D (N = 933,970), BMI (N = 806,834), and two GM datasets from the international consortium MiBioGen (211 taxa, N = 18,340) and the Dutch Microbiome Project (DMP) (207 taxa, N = 7,738). These datasets mainly cover European populations, with additional cohorts from Asia and other regions. To further explore the potential mediating role of GM in the connections between BMI and T2D, their interaction patterns were summarized into a network. RESULTS MR analysis identified 9 taxa that showed protective properties against T2D. Seven species were within the Firmicutes and Bacteroidales phyla in the DMP, and two were from the MiBioGen (Odds Ratio (OR): 0.94-0.95). Conversely, genetic components contributing to the abundance of 12 taxa were associated with increased risks of T2D (OR: 1.04-1.12). Furthermore, T2D may elevate the abundance of seven taxa (OR: 1.03-1.08) and reduce the abundance of six taxa (OR: 0.93-0.97). In the analysis of the influence of the genetic component of BMI on GM composition, BMI affected 52 bacterial taxa, with 28 decreasing (OR: 0.75-0.92) and 24 increasing (OR: 1.08-1.27). Besides, abundances of 25 taxa were negatively correlated with BMI (OR: 0.95-0.99), while positive correlations were detected for 14 taxa (OR: 1.01-1.05). Notably, we uncovered 11 taxa genetically associated with both BMI and T2D, which formed an interactive network. CONCLUSIONS Our findings provide evidence for the GM-mediated links between obesity and T2D. The identification of relevant GM taxa offers valuable insights into the potential role of the microbiome in these diseases.
Collapse
Affiliation(s)
- Li Fu
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
- Research Centre for Medical Genetics, Moscow, 115478, Russia
| | - Hongbao Cao
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
| | - Fuquan Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
5
|
Thonusin C, Suparan K, Kunasol C, Lungruammit N, Nawara W, Arunsak B, Kerdphoo S, Kongkaew A, Songtrai S, Pintana H, Maneechote C, Pratchayasakul W, Kaewsuwan S, Chattipakorn N, Chattipakorn SC. Interruptins Extracted from Cyclosorus terminans Protect Gut Pathologies Induced by High-Fat Diet in Rats. Nutrients 2025; 17:1387. [PMID: 40284250 PMCID: PMC12030309 DOI: 10.3390/nu17081387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES The fern "Cyclosorus terminans" (C. terminans) or "Maiden Fern" contains interruptin A and interruptin B. This plant could attenuate obesity, insulin resistance, and fatty liver in rats fed a high-fat/calorie diet. However, the benefits of C. terminans to the gut remain unknown. We investigated the protective effect of C. terminans extract against gut dysfunction in rats exposed to a high-fat/calorie diet. METHODS Male Wistar rats were assigned to receive either (1) a normal diet treated with vehicle, (2) a high-fat/calorie diet treated with vehicle, (3) a high-fat/calorie diet treated with 100 mg per kg per day (mg·kg-1·day-1) of C. terminans extract, or (4) a high-fat/calorie diet treated with 200 mg·kg-1·day-1 of C. terminans extract. The rats were euthanized after 12 weeks of treatment to enable feces and colon tissue collection. RESULTS Both 100 and 200 mg·kg-1·day-1 of C. terminans extract reduced body weight (-10.49%; p = 0.030 and -10.54%; p = 0.037, respectively) and ameliorated gut inflammation, gut barrier disruption, changes in short-chain fatty acid levels, and gut dysbiosis caused by high-fat/calorie diet. CONCLUSIONS C. terminans extract attenuated an increase in body weight and exerted prophylactic effects against gut pathologies induced by high-fat/calorie diet.
Collapse
Affiliation(s)
- Chanisa Thonusin
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanokphong Suparan
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chanon Kunasol
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Wichwara Nawara
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Aphisek Kongkaew
- Research Administration Section, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Sujinda Songtrai
- Faculty of Medical Technology, Rangsit University, Pathumthani 12000, Thailand;
| | - Hiranya Pintana
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wasana Pratchayasakul
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sireewan Kaewsuwan
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkhla University, Songkhla 90110, Thailand;
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkhla University, Songkhla 90110, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
6
|
Okamura T, Hasegawa Y, Ohno Y, Saijo Y, Nakanishi N, Honda A, Hamaguchi M, Takano H, Fukui M. Oral exposure to nanoplastics and food allergy in mice fed a normal or high-fat diet. CHEMOSPHERE 2025; 379:144401. [PMID: 40252413 DOI: 10.1016/j.chemosphere.2025.144401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
The global prevalence of food allergies, particularly IgE-mediated responses, is increasing at an alarming rate. This trend is likely driven by environmental factors such as nanoplastics (NPs) ingestion and the westernization of dietary and lifestyle habits. This study examines the impact of polystyrene nanoplastics (PS-NPs) on ovalbumin (OVA)-induced food allergies in mice subjected to either a normal diet (ND) or a high-fat diet (HFD). BALB/c mice were stratified into eight groups based on dietary regimen, NP exposure, and OVA sensitization. Food allergy was induced via OVA administration, and multiple physiological and immunological parameters were evaluated, including body weight, intestinal permeability, cytokine profiles, gut microbiota composition, and small intestinal gene expression. Mice in the HFD + OVA + NP group exhibited significant increases in intestinal permeability, diarrhea severity, and serum OVA-specific IgE levels compared to other groups. Flow cytometric analysis revealed an expansion of innate lymphoid cells (ILC2 and ILC1) within the lamina propria of the small intestine. Shotgun metagenomic sequencing demonstrated gut microbiota dysbiosis, characterized by a reduction in beneficial bacterial populations in the HFD + OVA + NP cohort. Weighted Gene Co-Expression Network Analysis (WGCNA) identified a negative correlation between NPs exposure or OVA sensitization and the expression of Slc1a1, Slc5a8, and Mep1a, while a positive correlation was observed with Aa467197 expression. These findings indicate that oral exposure to PS-NPs exacerbates OVA-induced food allergies, particularly in the context of an HFD, through mechanisms involving increased intestinal permeability, gut microbial dysbiosis, and gene expression modulation. This study highlights the potential health hazards posed by environmental microplastic contamination and its possible contribution to the escalating incidence of food allergies.
Collapse
Affiliation(s)
- Takuro Okamura
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, 602-8566, Japan
| | - Yuka Hasegawa
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, 602-8566, Japan
| | - Yuriko Ohno
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, 602-8566, Japan
| | - Yuto Saijo
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, 602-8566, Japan
| | - Naoko Nakanishi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, 602-8566, Japan
| | - Akiko Honda
- Environmental Health Sciences, Graduate School of Global Environmental Studies, Kyoto University, Kyoto, 615-8530, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, 602-8566, Japan.
| | - Hirohisa Takano
- Environmental Health Sciences, Graduate School of Global Environmental Studies, Kyoto University, Kyoto, 615-8530, Japan; Kyoto University of Advanced Science, Kyoto, 615-8577, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, 602-8566, Japan
| |
Collapse
|
7
|
Mishra S, Jain S, Agadzi B, Yadav H. A Cascade of Microbiota-Leaky Gut-Inflammation- Is it a Key Player in Metabolic Disorders? Curr Obes Rep 2025; 14:32. [PMID: 40208464 DOI: 10.1007/s13679-025-00624-0] [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] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE OF REVIEW This review addresses critical gaps in knowledge and provides a literature overview of the molecular pathways connecting gut microbiota dysbiosis to increased intestinal permeability (commonly referred to as "leaky gut") and its contribution to metabolic disorders. Restoring a healthy gut microbiota holds significant potential for enhancing intestinal barrier function and metabolic health. These interventions offer promising therapeutic avenues for addressing leaky gut and its associated pathologies in metabolic syndrome. RECENT FINDINGS In metabolic disorders such as obesity and type 2 diabetes (T2D), beneficial microbes such as those producing short-chain fatty acids (SCFAs) and other key metabolites like taurine, spermidine, glutamine, and indole derivatives are reduced. Concurrently, microbes that degrade toxic metabolites such as ethanolamine also decline, while proinflammatory, lipopolysaccharide (LPS)-enriched microbes increase. These microbial shifts place a higher burden on intestinal epithelial cells, which are in closest proximity to the gut lumen, inducing detrimental changes that compromise the structural and functional integrity of the intestinal barrier. Such changes include exacerbation of tight junction protein (TJP)s dysfunction, particularly through mechanisms such as destabilization of zona occludens (Zo)-1 mRNA or post-translational modifications. Emerging therapeutic strategies including ketogenic and Mediterranean diets, as well as probiotics, prebiotics, synbiotics, and postbiotics have demonstrated efficacy in restoring beneficial microbial populations, enhancing TJP expression and function, supporting gut barrier integrity, reducing leaky gut and inflammation, and ultimately improving metabolic disorders. This review summarizes the mechanisms by which gut microbiota contribute to the development of leaky gut and inflammation associated with metabolic syndrome. It also explores strategies for restoring gut microbiota balance and functionality by promoting beneficial microbes, increasing the production of beneficial metabolites, clearing toxic metabolites, and reducing the proportion of proinflammatory microbes. These approaches can alleviate the burden on intestinal epithelial cells, reduce leaky gut and inflammation, and improve metabolic health.
Collapse
Affiliation(s)
- Sidharth Mishra
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Bryan Agadzi
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Director of USF Center for Microbiome Research, Microbiomes Institute, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, MDC78, Tampa, FL, 33612, USA.
| |
Collapse
|
8
|
Hong Y, Yang M, Xu X, Wang P, Ten Z, Chen H, Fu M, Xiong R, Ouyang J. Gut microbiota, inflammatory proteins and bone mineral density in different age groups: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41875. [PMID: 40193639 PMCID: PMC11977711 DOI: 10.1097/md.0000000000041875] [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: 08/28/2024] [Revised: 12/12/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
Several studies have indicated a potential association between gut microbiota and bone density. However, the causal relationship between gut microbiota and bone mineral density across different age groups, as well as the potential role of inflammatory proteins as mediators, remains unclear. Gut microbiota, inflammatory proteins, and bone mineral density (BMD) were identified in various age groups using summary data from large-scale genome-wide association studies. Mendelian randomization was employed to examine the causal connections between gut microbiota, inflammatory proteins, and BMD in different age groups, primarily utilizing inverse variance weighted as the statistical method. Furthermore, the potential role of inflammatory proteins as mediators in the pathway from gut microbiota to BMD was investigated. Eight positive and 19 negative causal relationships between gut microbiota and BMD were observed across various age groups. We also identified 14 positive and 8 negative causal relationships between inflammatory proteins and BMD in different age groups. Inflammatory proteins did not appear to function as mediators in the pathway from gut microbiota to BMD. Gut microbiota and inflammatory proteins were causally linked to BMD; however, inflammatory proteins did not seem to function as mediators in the pathway from gut microbiota to BMD because the effects of intestinal flora on bone density and the effects of inflammatory factors on bone density were in different directions.
Collapse
Affiliation(s)
- Yuechang Hong
- School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People’s Republic of China
| | - Minghui Yang
- School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People’s Republic of China
| | - Xin Xu
- Department of Sports Medicine, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China
| | - Peng Wang
- Department of Sports Medicine, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China
| | - Zixin Ten
- School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People’s Republic of China
| | - Huang Chen
- School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People’s Republic of China
| | - Minqiang Fu
- School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, People’s Republic of China
| | - Renying Xiong
- Department of Medicine, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China
| | - Jianjiang Ouyang
- Department of Sports Medicine, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China
| |
Collapse
|
9
|
Mo X, Cheng R, Shen L, Sun Y, Wang P, Jiang G, Wen L, Li X, Peng X, Liao Y, He R, Yan H, Liu L. High-fat diet induces sarcopenic obesity in natural aging rats through the gut-trimethylamine N-oxide-muscle axis. J Adv Res 2025; 70:405-422. [PMID: 38744403 PMCID: PMC11976414 DOI: 10.1016/j.jare.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024] Open
Abstract
INTRODUCTION The lack of suitable animal models for sarcopenic obesity (SO) limits in-depth research into the disease. Emerging studies have demonstrated that gut dysbiosis is involved in the development of SO. As the importance of microbial metabolites is starting to unveil, it is necessary to comprehend the specific metabolites associated with gut microbiota and SO. OBJECTIVES We aimed to investigate whether high-fat diet (HFD) causes SO in natural aging animal models and specific microbial metabolites that are involved in linking HFD and SO. METHODS Young rats received HFD or control diet for 80 weeks, and obesity-related metabolic disorders and sarcopenia were measured. 16S rRNA sequencing and non-targeted and targeted metabolomics methods were used to detect fecal gut microbiota and serum metabolites. Gut barrier function was evaluated by intestinal barrier integrity and intestinal permeability. Trimethylamine N-oxide (TMAO) treatment was further conducted for verification. RESULTS HFD resulted in body weight gain, dyslipidemia, impaired glucose tolerance, insulin resistance, and systemic inflammation in natural aging rats. HFD also caused decreases in muscle mass, strength, function, and fiber cross-sectional area and increase in muscle fatty infiltration in natural aging rats. 16S rRNA sequencing and nontargeted and targeted metabolomics analysis indicated that HFD contributed to gut dysbiosis, mainly characterized by increases in deleterious bacteria and TMAO. HFD destroyed intestinal barrier integrity and increased intestinal permeability, as evaluated by reducing levels of colonic mucin-2, tight junction proteins, goblet cells and elevating serum level of fluorescein isothiocyanate-dextran 4. Correlation analysis showed a positive association between TMAO and SO. In addition, TMAO treatment aggravated the development of SO in HFD-fed aged rats through regulating the ROS-AKT/mTOR signaling pathway. CONCLUSION HFD leads to SO in natural aging rats, partially through the gut-microbiota-TMAO-muscle axis.
Collapse
Affiliation(s)
- Xiaoxing Mo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Ruijie Cheng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Lihui Shen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Yunhong Sun
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Pei Wang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Guanhua Jiang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Lin Wen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Xiaoqin Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Xiaobo Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Ruikun He
- CAS Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Hong Yan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
| |
Collapse
|
10
|
Chen J, Luo M, Xing Z, Chen Y, Peng C, Li D. Start small, think big: MicroRNAs in diabetes mellitus and relevant cardiorenal-liver metabolic health spectrum. Metabolism 2025; 165:156153. [PMID: 39914482 DOI: 10.1016/j.metabol.2025.156153] [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: 08/29/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Diabetes mellitus (DM), co-existing with metabolic disorder of cardio-renal-liver, is one of the most difficult problems in medicine that attracts global concern with high mortality. MicroRNAs (miRNAs) are a class of small non-coding RNA molecules that negatively regulates gene expression and exerts active against a large proportion of the transcriptome, due to their high evolutionary conservation. Emerging evidence prove that miRNAs are involved in the pathogenesis of DM and associated metabolic disorders, manifested by their variable alteration in the blood, urine, tissues, or organs, principally contributing to modulate the interconnections between DM and cardio-renal-liver metabolism. Mechanistically, miRNAs regulate various biological processes, such as metabolism of insulin, lipid, glucose, inflammatory response, fibrosis, oxidative stress, apoptosis, and angiogenesis, etc. This review emphasizes the function of miRNAs and highlights the physiopathological regulation of miRNA in DM and related complications, especially the dysfunction of cardiovascular system, kidneys, and liver, with the aim of providing promising biomarkers for assisting early diagnosis of DM with cardio-renal-liver- specific metabolic disorders, as well as for the development of miRNA-targeting agents.
Collapse
Affiliation(s)
- Junren Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maozhu Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziwei Xing
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
11
|
Li W, Zeng Y, Zhong J, Hu Y, Xiong X, Zhou Y, Fu L. Probiotics Exert Gut Immunomodulatory Effects by Regulating the Expression of Host miRNAs. Probiotics Antimicrob Proteins 2025; 17:557-568. [PMID: 39754704 DOI: 10.1007/s12602-024-10443-9] [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] [Accepted: 12/19/2024] [Indexed: 01/06/2025]
Abstract
Probiotics exert a diverse range of immunomodulatory effects on the human gut immune system. These mechanisms encompass strengthening the intestinal mucosal barrier, inhibiting pathogen adhesion and colonization, stimulating immune modulation, and fostering the production of beneficial substances. As a result, probiotics hold significant potential in the prevention and treatment of various conditions, including inflammatory bowel disease and colorectal cancer. A pivotal mechanism by which probiotics achieve these effects is through modulating the expression of host miRNAs. miRNAs, non-coding RNA molecules, are vital regulators of fundamental biological processes like cell growth, differentiation, and apoptosis. By interacting with mRNAs, miRNAs can either promote their degradation or repress their translation, thereby regulating gene expression post-transcriptionally and modulating the immune system. This review provides a comprehensive overview of how probiotics modulate gut immune responses by altering miRNA expression levels, both upregulating and downregulating specific miRNAs. It further delves into how this modulation impacts the host's resistance to pathogens and susceptibility to diseases, offering a theoretical foundation and practical insights for the clinical utilization of probiotics in disease prevention and therapy.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Yongwei Zeng
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Jiayu Zhong
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Youyu Hu
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Xia Xiong
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yingshun Zhou
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
- Public Center of Experimental Technology of Pathogen Biology Technology Platform, Southwest Medical University, Luzhou, 646000, China.
| | - Li Fu
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
12
|
Zhao YY, Wu ZJ, Du Y, Han QQ, Bai YY, Liu B, Li J. Gut microbiome and serum metabolites in neuropathic pain: The PPARα perspective. Behav Brain Res 2025; 482:115442. [PMID: 39864460 DOI: 10.1016/j.bbr.2025.115442] [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: 12/03/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
Neuropathic pain (NP) is a chronic disease state centred on neuroinflammation with a high prevalence and limited effective treatment options. Peroxisome proliferator-activated receptor α (PPARα) has emerged as a promising target for NP management due to its anti-inflammatory properties. Recent evidence highlights the critical role of the gut microbiome and its metabolites in NP pathogenesis. This study aimed to investigate whether PPARα modulates the development and alleviation of NP by influencing gut microbial communities and serum metabolites. 16S rDNA sequencing and liquid chromatography-mass spectrometry (LC-MS/MS) untargeted metabolomics analyses performed 14 days after the establishment of a chronic constriction injury (CCI) pain model in C57BL/6 J mice showed significant changes in gut microbial and metabolite levels in CCI mice. Intraperitoneal injection of the PPARα agonist GW7647 (5 mg/kg) significantly attenuated mechanical allodynia and thermal hyperalgesia in CCI mice, whereas injection of the PPARα antagonist GW6471 (20 mg/kg) produced the opposite effect. Immunofluorescence analysis revealed that GW7647 effectively suppressed microglial activation. Additionally, PPARα agonist and antagonist treatments markedly altered the composition and abundance of intestinal microbial communities in CCI mice. Further serum LC-MS/MS analysis identified 258 potential serum metabolic biomarkers, many of which correlated with changes in gut microbial composition. These findings demonstrate that PPARα influences serum metabolite profiles by modulating gut microbiota composition, which subsequently affects NP progression. This study provides novel insights into the mechanisms underlying NP and suggests potential therapeutic avenues targeting PPARα and gut microbiota.
Collapse
Affiliation(s)
- Yu-Ying Zhao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Zi-Jun Wu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yue Du
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Qing-Qing Han
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yuan-Yuan Bai
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Bin Liu
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Center for Critical Care Medicine, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300020, China.
| | - Jing Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| |
Collapse
|
13
|
Liu Y, Li X, Chen Y, Yao Q, Zhou J, Wang X, Meng Q, Ji J, Yu Z, Chen X. Fecal microbiota transplantation: application scenarios, efficacy prediction, and factors impacting donor-recipient interplay. Front Microbiol 2025; 16:1556827. [PMID: 40201444 PMCID: PMC11975908 DOI: 10.3389/fmicb.2025.1556827] [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: 01/07/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Fecal microbiota transplantation (FMT) represents a therapeutic approach that directly regulates the gut microbiota of recipients, normalizes its composition and reaping therapeutic rewards. Currently, in addition to its general application in treating Clostridium difficile (C. difficile) infection (CDI), FMT treatment has also been extended to the fields of other gastrointestinal diseases, infections, gut-liver or gut-brain axis disorders, metabolic diseases and cancer, etc. Prior to FMT, rigorous donor screening is essential to reduce the occurrence of adverse events. In addition, it is imperative to evaluate whether the recipient can safely and effectively undergo FMT treatment. However, the efficacy of FMT is influenced by the complex interactions between the gut microbiota of donor and recipient, the degree of donor microbiota engraftment is not necessarily positively related with the success rate of FMT. Furthermore, an increasing number of novel factors affecting FMT outcomes are being identified in recent clinical trials and animal experiments, broadening our understanding of FMT treatment. This article provides a comprehensive review of the application scenarios of FMT, the factors influencing the safety and efficacy of FMT from the aspects of both the donors and the recipients, and summarizes how these emerging novel regulatory factors can be combined to predict the clinical outcomes of patients undergoing FMT.
Collapse
Affiliation(s)
- Yaxin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinru Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuchao Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qinyan Yao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinjie Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoxuan Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qingguo Meng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiaxuan Ji
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
14
|
Haque PS, Goodman D, Kuusivuori-Robinson T, Coughlan C, Delgado-Deida Y, Onyiah JC, Zempleni J, Theiss AL. Obese Adipose Tissue Extracellular Vesicles Activate Mitochondrial Fatty Acid β-oxidation to Drive Colonic Stemness. Cell Mol Gastroenterol Hepatol 2025; 19:101504. [PMID: 40122519 DOI: 10.1016/j.jcmgh.2025.101504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND & AIMS Patients with obesity and mouse models of obesity exhibit abnormalities in intestinal epithelial cells, including enhanced stemness. Adipose tissue (AT) is the largest endocrine organ secreting cytokines, hormones, and extracellular vesicles (EVs). Here, we characterized EV protein cargo from obese and non-obese AT and demonstrate the role of obese adipose-derived EVs in enhancing colonic stemness. METHODS EVs were isolated from visceral AT from mice fed high-fat diet to induce obesity or control matched-diet. EV cargo was characterized by unbiased proteomics. Mouse colonoids were treated with EVs and analyzed for fatty acid β-oxidation (FAO), expression of stem marker genes, stem function, and β-catenin expression and acetylation. Mice deficient in adipocyte-specific Tsg101 expression were generated to alter adipocyte EV protein cargo, and colonic stemness was measured. RESULTS EVs secreted from obese visceral AT (Ob EVs) were significantly enriched with acyl-CoA dehydrogenase long chain (ACADL), an initiator enzyme of FAO. Compared with non-obese EVs, colonoids treated with Ob EVs exhibited increased exogenous ACADL protein expression, FAO, growth, persistence of stem/progenitor function, and increased β-catenin protein expression and acetylation that was abolished by FAO inhibition. Mice deficient in adipocyte-specific Tsg101 expression exhibited Ob EVs with altered protein expression profiles and were protected from obesity-induced enhanced colonic stemness. CONCLUSIONS The contents of Ob EVs are poised to fuel FAO and to promote obesity-induced stemness in the colon. Alteration of metabolism is a key mechanism of adipose-to-intestinal tissue communication elicited by EVs, thereby influencing basal colonic stem cell homeostasis during obesity.
Collapse
Affiliation(s)
- Parsa S Haque
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Desiree Goodman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Thor Kuusivuori-Robinson
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Christina Coughlan
- Division of Neurology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Yaritza Delgado-Deida
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Joseph C Onyiah
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Arianne L Theiss
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado.
| |
Collapse
|
15
|
Zhu D, Li S, Xu Z, Kulyar MF, Bai X, Wang Y, Wang B, Khateeb E, Deng D, Wang L, Chen Y, Guo A, Shen Y. Comparative analysis of gut microbiota in healthy and diarrheic foals. Microbiol Spectr 2025:e0087124. [PMID: 40105330 DOI: 10.1128/spectrum.00871-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 02/01/2025] [Indexed: 03/20/2025] Open
Abstract
Diarrhea presents a substantial risk of high morbidity and mortality among foals. Although studies have shown connections between gut microbiota and several gastrointestinal diseases, there is still inadequate information on gut microbial alterations in foals during diarrhea. In this study, we conducted 16S rRNA and ITS gene amplicon sequencing to investigate gut bacterial and fungal differences between healthy and diarrheic foals. The results unveiled significant reductions in gut bacterial and fungal diversities among foals experiencing diarrhea, accompanied by notable shifts in the composition of gut microbial communities. A considerable decrease was observed in the relative abundance of 30 bacterial and 34 fungal genera. Moreover, two bacterial and eight fungal genera were utterly undetectable in the gut microbiota of diarrheic foals. Some decreased genera, such as Bifidobacterium and Saccharomyces, were deemed beneficial and recognized as probiotics. The study revealed significant alterations in foals' gut bacterial and fungal communities during diarrhea, which enriched our comprehension of gut microbial dynamics in foals across varying health statuses. These findings offer valuable insights for managing diarrhea through gut microbiota modulation, suggesting that probiotics may be superior to antibiotics in preventing and controlling foal diarrhea.IMPORTANCEThis research advances the understanding of gut bacterial and fungal dynamics in foals, highlighting gut microbiota dysbiosis as a potential contributor to foal diarrhea. Additionally, we observed that many altered bacteria and fungi were downregulated during diarrhea, including some probiotic strains. Consequently, our findings provide evidence that probiotics may offer superior efficacy compared with antibiotics as potential candidates for preventing and treating foal diarrhea.
Collapse
Affiliation(s)
- Di Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Siyu Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhixiang Xu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Md F Kulyar
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xu Bai
- China Horse Industry Association, Beijing, China
| | - Yu Wang
- China Horse Industry Association, Beijing, China
| | - Boya Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Emaan Khateeb
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Dandan Deng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lidan Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yuji Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Aizhen Guo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yaoqin Shen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
16
|
Li J, Zhang YJ, Zhao X, Yu Y, Xu JH, Hu R, Wu YH, Huang WQ, Wang ZX, Li TT. Impact of sodium butyrate on stroke-related intestinal injury in diabetic mice: Interference with Caspase-1/GSDMD pyroptosis pathway and preservation of intestinal barrier. Eur J Pharmacol 2025; 998:177455. [PMID: 40057153 DOI: 10.1016/j.ejphar.2025.177455] [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: 07/15/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
Diabetic stroke-associated acute intestinal injury is characterized by high mortality, disability, and poor prognosis due to the lack of effective therapies. Our prior research demonstrated that administration of 300 mg/kg sodium butyrate (NaB) can improve neurological outcomes post-diabetic stroke. Nonetheless, whether the effect of NaB is related to intestinal regulation, along with its underlying mechanisms, remains uncertain. This study aims to investigate the effects and mechanistic pathways of NaB on diabetic stroke-associated acute intestinal injury. A middle cerebral artery occlusion/reperfusion model was established in mice with streptozotocin-induced diabetes. The results demonstrated that NaB alleviated colonic injury 24 h after reperfusion in diabetic stroke. Pyroptosis-related protein levels in colonic tissues were significantly elevated following diabetic stroke but were markedly reduced with NaB treatment. NaB also improved gut barrier integrity and reduced inflammation, promoting epithelial barrier self-repair. In the NaB combined with lipopolysaccharide group, lipopolysaccharide administration induced a significant inflammatory response in the colonic tissue. Conversely, treatment with NaB and VX-765 (an inhibitor for Caspase-1) led to a notable alleviation in intestinal inflammation. These findings suggest that NaB mitigates colonic injury and enhances barrier function following diabetic stroke, potentially through the Caspase-1/Gasdermin D pyroptosis pathway. This study may provide a novel strategy and direction for intestinal rehabilitation in diabetic stroke patients.
Collapse
Affiliation(s)
- Jing Li
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuan-Jia Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xu Zhao
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Yu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing-Hong Xu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ye-Hui Wu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wen-Qi Huang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Zhong-Xing Wang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Ting-Ting Li
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
17
|
Thiran A, Vereecke L. New thoughts on the gut-immune axis of arthritis. Trends Immunol 2025; 46:206-218. [PMID: 40069048 DOI: 10.1016/j.it.2025.01.006] [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: 11/20/2024] [Revised: 12/23/2024] [Accepted: 01/27/2025] [Indexed: 03/21/2025]
Abstract
Arthritis is associated with varying degrees of intestinal inflammation and microbiota dysbiosis, leading to the 'gut-joint axis hypothesis' in which intestinal and joint inflammation are suggested to be interconnected through immune-microbiota interactions. While clinical observations support this, causality remains uncertain. Rodent models have provided insights into potential mechanisms by uncovering microbial influences and immune pathways that either connect or uncouple gut and joint inflammation. Based on recent findings, we propose the 'immune hypersensitivity hypothesis' whereby central immune hyper-reactivity can independently drive joint inflammation via local sterile triggers, and gut inflammation via microbial triggers. We argue that this suggests a more nuanced role of the microbiota in arthritis pathogenesis that varies according to the predominant immune mechanisms in disease subtypes. We explore gut-immune interactions in arthritis, highlight ongoing challenges, and propose future research directions.
Collapse
Affiliation(s)
- Alexandra Thiran
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Vlaams Instituut voor Biotechnologie (VIB)-University of Ghent (UGent) Center for Inflammation Research, Ghent, Belgium; Ghent Gut Inflammation Group (GGIG), Ghent, Belgium
| | - Lars Vereecke
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Vlaams Instituut voor Biotechnologie (VIB)-University of Ghent (UGent) Center for Inflammation Research, Ghent, Belgium; Ghent Gut Inflammation Group (GGIG), Ghent, Belgium.
| |
Collapse
|
18
|
Li X, Lin D, Hu X, Shi X, Huang W, Ouyang Y, Chen X, Xiong Y, Wu X, Hong D, Chen H. Akkermansia muciniphila Modulates Central Nervous System Autoimmune Response and Cognitive Impairment by Inhibiting Hippocampal NLRP3-Mediated Neuroinflammation. CNS Neurosci Ther 2025; 31:e70320. [PMID: 40050112 PMCID: PMC11884925 DOI: 10.1111/cns.70320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 01/20/2025] [Accepted: 02/11/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Numerous studies have demonstrated the significant role of Akkermansia muciniphila (A. muciniphila) in enhancing host immune responses and metabolic functions. However, its increased presence in multiple sclerosis (MS) patients has led to a focus on the relationships between A. muciniphila and diseases, with the underlying mechanisms remaining unknown. METHOD Solochrome cyanin, hematoxylin-eosin staining (H&E) and immunofluorescence staining were used to assess demyelination and inflammation. Gut microbiota changes were examined by 16S rRNA sequencing. Intracellular cytokine levels were assessed by flow cytometry. Cognitive impairment was evaluated using four behavioral tests. Intestinal barrier function and pyrin domain-containing protein 3 (NLRP3)-mediated neuroinflammation were evaluated by immunoblotting. RESULTS We found that treatment with an appropriate dose of A. muciniphila (5.0 × 107 CFU/mL) reduced neuropathology and disease severity in experimental autoimmune encephalomyelitis (EAE) mice. In addition, A. muciniphila supplementation increased the diversity and abundance of intestinal microbiota while decreasing the Firmicutes/Bacteroidetes ratio. Moreover, it improved intestinal barrier function and attenuated Th17 responses in the gut, central nervous system (CNS), and lymphoid tissues, without affecting Treg response in the lymphoid tissue. Furthermore, A. muciniphila administration partly regulated cognitive impairment and hippocampal NLRP3-mediated neuroinflammation. CONCLUSION Our results suggest that A. muciniphila holds promise as a probiotic for treating NLRP3-associated inflammatory disorders and cognitive impairment, including MS.
Collapse
Affiliation(s)
- Xiaobing Li
- Department of NeurologyThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Institute of Neurology, Jiangxi Academy of Clinical Medical Science, the First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health CommissionJiangxi Medical College, Nanchang UniversityNanchangChina
| | - Dengna Lin
- Department of GastroenterologyThe Sixth Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
- Guangdong Institute of GastroenterologyGuangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical DisciplineGuangzhouChina
| | - Xin Hu
- Department of NeurologyThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Institute of Neurology, Jiangxi Academy of Clinical Medical Science, the First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health CommissionJiangxi Medical College, Nanchang UniversityNanchangChina
| | - Xiongwei Shi
- Department of NeurologyThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Institute of Neurology, Jiangxi Academy of Clinical Medical Science, the First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health CommissionJiangxi Medical College, Nanchang UniversityNanchangChina
| | - Wenxuan Huang
- Department of NeurologyThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Institute of Neurology, Jiangxi Academy of Clinical Medical Science, the First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health CommissionJiangxi Medical College, Nanchang UniversityNanchangChina
| | - Yi Ouyang
- Department of NeurologyThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Institute of Neurology, Jiangxi Academy of Clinical Medical Science, the First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health CommissionJiangxi Medical College, Nanchang UniversityNanchangChina
| | - Xiaohong Chen
- Department of Neurology and Multiple Sclerosis Research CenterThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yingqiong Xiong
- Department of NeurologyThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Institute of Neurology, Jiangxi Academy of Clinical Medical Science, the First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health CommissionJiangxi Medical College, Nanchang UniversityNanchangChina
| | - Xiaomu Wu
- Department of NeurologyThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Institute of Neurology, Jiangxi Academy of Clinical Medical Science, the First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health CommissionJiangxi Medical College, Nanchang UniversityNanchangChina
| | - Daojun Hong
- Department of NeurologyThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Institute of Neurology, Jiangxi Academy of Clinical Medical Science, the First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health CommissionJiangxi Medical College, Nanchang UniversityNanchangChina
| | - Hao Chen
- Department of NeurologyThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Institute of Neurology, Jiangxi Academy of Clinical Medical Science, the First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
- Key Laboratory of Rare Neurological Diseases of Jiangxi Provincial Health CommissionJiangxi Medical College, Nanchang UniversityNanchangChina
| |
Collapse
|
19
|
Barra NG, Fang H, Bhatwa A, Schmidt AM, Syed SA, Steinberg GR, Morrison KM, Surette MG, Wade MG, Holloway AC, Schertzer JD. Food supply toxicants and additives alter the gut microbiota and risk of metabolic disease. Am J Physiol Endocrinol Metab 2025; 328:E337-E353. [PMID: 39871724 DOI: 10.1152/ajpendo.00364.2024] [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: 09/17/2024] [Revised: 10/09/2024] [Accepted: 01/13/2025] [Indexed: 01/29/2025]
Abstract
Metabolic disease is rising along with both global industrialization and the use of new commercial, agricultural, and industrial chemicals and food additives. Exposure to these compounds may contribute to aspects of metabolic diseases such as obesity, diabetes, and fatty liver disease. Ingesting compounds in the food supply is a key route of human exposure, resulting in the interaction between toxicants or additives and the intestinal microbiota. Toxicants can influence the composition and function of the gut microbiota, and these microbes can metabolize and transform toxicants and food additives. Microbe-toxicant interactions in the intestine can alter host mucosal barrier function, immunity, and metabolism, which may contribute to the risk or severity of metabolic disease development. Targeting the connection between toxicants, food, and immunity in the gut using strategies such as fermentable fiber (i.e., inulin) may mitigate some of the effects of these compounds on host metabolism. Understanding causative factors in the microbe-host relationship that promote toxicant-induced dysmetabolism is an important goal. This review highlights the role of common toxicants (i.e., persistent organic pollutants, pesticides, and fungicides) and food additives (emulsifiers and artificial sweeteners) found in our food supply that alter the gut microbiota and promote metabolic disease development.
Collapse
Affiliation(s)
- Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Han Fang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Arshpreet Bhatwa
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Angela M Schmidt
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Saad A Syed
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Katherine M Morrison
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Michael G Wade
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Alison C Holloway
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
20
|
Tadese DA, Mwangi J, Luo L, Zhang H, Huang X, Michira BB, Zhou S, Kamau PM, Lu Q, Lai R. The microbiome's influence on obesity: mechanisms and therapeutic potential. SCIENCE CHINA. LIFE SCIENCES 2025; 68:657-672. [PMID: 39617855 DOI: 10.1007/s11427-024-2759-3] [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: 06/21/2024] [Accepted: 10/16/2024] [Indexed: 01/03/2025]
Abstract
In 2023, the World Obesity Atlas Federation concluded that more than 50% of the world's population would be overweight or obese within the next 12 years. At the heart of this epidemic lies the gut microbiota, a complex ecosystem that profoundly influences obesity-related metabolic health. Its multifaced role encompasses energy harvesting, inflammation, satiety signaling, gut barrier function, gut-brain communication, and adipose tissue homeostasis. Recognizing the complexities of the cross-talk between host physiology and gut microbiota is crucial for developing cutting-edge, microbiome-targeted therapies to address the global obesity crisis and its alarming health and economic repercussions. This narrative review analyzed the current state of knowledge, illuminating emerging research areas and their implications for leveraging gut microbial manipulations as therapeutic strategies to prevent and treat obesity and related disorders in humans. By elucidating the complex relationship between gut microflora and obesity, we aim to contribute to the growing body of knowledge underpinning this critical field, potentially paving the way for novel interventions to combat the worldwide obesity epidemic.
Collapse
Affiliation(s)
- Dawit Adisu Tadese
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - James Mwangi
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lei Luo
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Zhang
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Xiaoshan Huang
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Brenda B Michira
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shengwen Zhou
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peter Muiruri Kamau
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiumin Lu
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Ren Lai
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
21
|
Marette A, Pilon G. Host-microbiome determinants of insulin resistance in obesity: alone we go faster, together we go further! Gut 2025:gutjnl-2024-333855. [PMID: 39933914 DOI: 10.1136/gutjnl-2024-333855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Affiliation(s)
- Andre Marette
- Institute of Nutrition and Functional Foods, Laval University, Quebec, Canada
- Department of Medicine and IUCPQ, Laval University, Quebec, Quebec, Canada
| | - Genevieve Pilon
- Institute of Nutrition and Functional Foods, Laval University, Quebec, Canada
| |
Collapse
|
22
|
Nai S, Song J, Su W, Liu X. Bidirectional Interplay Among Non-Coding RNAs, the Microbiome, and the Host During Development and Diseases. Genes (Basel) 2025; 16:208. [PMID: 40004537 PMCID: PMC11855195 DOI: 10.3390/genes16020208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
It is widely known that the dysregulation of non-coding RNAs (ncRNAs) and dysbiosis of the gut microbiome play significant roles in host development and the progression of various diseases. Emerging evidence has highlighted the bidirectional interplay between ncRNAs and the gut microbiome. This article aims to review the current understanding of the molecular mechanisms underlying the crosstalk between ncRNAs, especially microRNA (miRNA), and the gut microbiome in the context of development and diseases, such as colorectal cancer, inflammatory bowel diseases, neurological disorders, obesity, and cardiovascular disease. Ultimately, this review seeks to provide a foundation for exploring the potential roles of ncRNAs and gut microbiome interactions as biomarkers and therapeutic targets for clinical diagnosis and treatment, such as ncRNA mimics, antisense oligonucleotides, and small-molecule compounds, as well as probiotics, prebiotics, and diets.
Collapse
Affiliation(s)
| | | | | | - Xiaoqian Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (S.N.); (J.S.); (W.S.)
| |
Collapse
|
23
|
Dou Y, Niu Y, Shen H, Wang L, Lv Y, Liu S, Xie X, Feng A, Liu X. Identification of disease-specific gut microbial markers in vitiligo. Front Microbiol 2025; 16:1499035. [PMID: 39967732 PMCID: PMC11833150 DOI: 10.3389/fmicb.2025.1499035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/21/2025] [Indexed: 02/20/2025] Open
Abstract
There is a potential correlation between vitiligo and gut microbiota, although research in this area is currently limited. The research employed high-throughput sequencing of 16S rRNA to examine the gut microbiome in the stool samples of 49 individuals with vitiligo and 49 without the condition. The study encompassed four comparison groups: (1) DI (disease) group vs. HC (healthy control) group; (2) DI_m group (disease group of minors) vs. HC_m group (healthy control group of minors); (3) DI_a group (adult disease group) vs. HC_a group (adult healthy control group); (4) DI_m group vs. DI_a group. Research findings have indicated the presence of spatial heterogeneity in the gut microbiota composition between individuals with vitiligo and healthy controls. A significant reduction in gut microbiota diversity has been observed in vitiligo patients across both minors and adult groups. However, variations have been noted in the composition of disease-related differential microbial markers among different age groups. Specifically, Bacteroides and Parabacteroides have been identified as specific markers of the intestinal microbiota of vitiligo patients in both minor and adult groups. Correlative analyses have revealed a positive correlation of these two genera with the Vitiligo Area Scoring Index (VASI) and disease duration. It is noteworthy that there are no significant differences in diversity between the DI_m group and the DI_a group, with similarities in microbiota composition and functional characteristics. Nevertheless, correlative analyses suggest a declining trend in Bacteroides and Parabacteroides with increasing age. Individuals with vitiligo exhibit distinct features in their gut microbiome when contrasted with those in the healthy control group. Additionally, the microbial marker genera that show variances between patients and healthy controls vary among different age groups. Disease-specific microbial marker genera (Bacteroides and Parabacteroides) are associated with VASI, duration of the condition, and age. These findings are essential for improving early diagnosis and developing potential treatment strategies for individuals with vitiligo.
Collapse
Affiliation(s)
- Yimin Dou
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Niu
- Department of Gastroenterology Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hexiao Shen
- School of Life Science, Hubei University, Wuhan, China
| | - Lan Wang
- School of Life Science, Hubei University, Wuhan, China
| | - Yongling Lv
- School of Life Science, Hubei University, Wuhan, China
| | - Suwen Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiafei Xie
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aiping Feng
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinxin Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
Jiang Y, Yang J, Wei M, Shou J, Shen S, Yu Z, Zhang Z, Cai J, Lyu Y, Yang D, Han Y, Zhu J, Liu Z, Ma D, Xing GG, Li M. Probiotics alleviate painful diabetic neuropathy by modulating the microbiota-gut-nerve axis in rats. J Neuroinflammation 2025; 22:30. [PMID: 39894793 PMCID: PMC11789326 DOI: 10.1186/s12974-025-03352-3] [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: 10/28/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025] Open
Abstract
Painful diabetic neuropathy (PDN) is one of the most common complications of diabetes. Recent studies suggested that gut microbiota dysbiosis contributes to the development of PDN, but underlying mechanisms remain elusive. In this study, we found decreased probiotics generating bacteria such as Lactobacillus and Bifidobacterium strains in the PDN rats. Supplementation with multiple probiotics for 12 weeks alleviated pain, reversed nerve fiber lesions, and restored neuronal hyperexcitability. Probiotics administration effectively attenuated intestinal barrier impairment, reduced serum lipopolysaccharide and proinflammatory cytokines, and mitigated disruptions in the blood-nerve barrier. Furthermore, probiotics treatment inhibited the activation of the TLR4/MyD88/NF-κB signaling pathway and reduced proinflammatory cytokines in the sciatic nerve of the PDN rats. Together, our findings suggest that gut microbiota dysbiosis participates in PDN pathogenesis, and probiotics offer therapeutic potential via modulating the microbiota-gut-nerve axis.
Collapse
Affiliation(s)
- Ye Jiang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jing Yang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Min Wei
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jiayin Shou
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Shixiong Shen
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Zhuoying Yu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Zixian Zhang
- Neuroscience Research Institute, Peking University, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing, China
| | - Jie Cai
- Neuroscience Research Institute, Peking University, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing, China
| | - Yanhan Lyu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Dongsheng Yang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yongzheng Han
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jinpiao Zhu
- Perioperative and Systems Medicine Laboratory, Department of Anesthesiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Zhigang Liu
- Perioperative and Systems Medicine Laboratory, Department of Anesthesiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Daqing Ma
- Perioperative and Systems Medicine Laboratory, Department of Anesthesiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK.
| | - Guo-Gang Xing
- Neuroscience Research Institute, Peking University, Beijing, China.
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing, China.
| | - Min Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
25
|
Lin X, Yu Z, Liu Y, Li C, Hu H, Hu J, Liu M, Yang Q, Gu P, Li J, Nandakumar KS, Hu G, Zhang Q, Chen X, Ma H, Huang W, Wang G, Wang Y, Huang L, Wu W, Liu N, Zhang C, Liu X, Zheng L, Chen P. Gut-X axis. IMETA 2025; 4:e270. [PMID: 40027477 PMCID: PMC11865426 DOI: 10.1002/imt2.270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 03/05/2025]
Abstract
Recent advances in understanding the modulatory functions of gut and gut microbiota on human diseases facilitated our focused attention on the contribution of the gut to the pathophysiological alterations of many extraintestinal organs, including the liver, heart, brain, lungs, kidneys, bone, skin, reproductive, and endocrine systems. In this review, we applied the "gut-X axis" concept to describe the linkages between the gut and other organs and discussed the latest findings related to the "gut-X axis," including the underlying modulatory mechanisms and potential clinical intervention strategies.
Collapse
Affiliation(s)
- Xu Lin
- Department of Endocrinology and MetabolismShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)Foshan City528308China
| | - Zuxiang Yu
- State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, The Institute of Cardiovascular Sciences and Institute of Systems BiomedicinePeking UniversityBeijing100191China
| | - Yang Liu
- State Key Laboratory of Reproductive Medicine and Offsprings Health, Center for Global HealthNanjing Medical UniversityNanjing211166China
| | - Changzhou Li
- Department of Plastic and Aesthetic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Hui Hu
- Department of Laboratory Medicine, Shanghai East HospitalTongji University School of MedicineShanghai200123China
| | - Jia‐Chun Hu
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Mian Liu
- Department of Obstetrics and Gynecology, Nanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Qin Yang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Peng Gu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Jiaxin Li
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Kutty Selva Nandakumar
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
| | - Gaofei Hu
- State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, The Institute of Cardiovascular Sciences and Institute of Systems BiomedicinePeking UniversityBeijing100191China
| | - Qi Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, The Institute of Cardiovascular Sciences and Institute of Systems BiomedicinePeking UniversityBeijing100191China
| | - Xinyu Chen
- State Key Laboratory of Reproductive Medicine and Offsprings Health, Center for Global HealthNanjing Medical UniversityNanjing211166China
| | - Huihui Ma
- State Key Laboratory of Reproductive Medicine and Offsprings Health, Center for Global HealthNanjing Medical UniversityNanjing211166China
| | - Wenye Huang
- State Key Laboratory of Reproductive Medicine and Offsprings Health, Center for Global HealthNanjing Medical UniversityNanjing211166China
| | - Gaofeng Wang
- Department of Plastic and Aesthetic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Yan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Liping Huang
- Department of Obstetrics and Gynecology, Nanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Wenjuan Wu
- Department of Laboratory Medicine, Shanghai East HospitalTongji University School of MedicineShanghai200123China
| | - Ning‐Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Chenhong Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghai200240China
| | - Xingyin Liu
- State Key Laboratory of Reproductive Medicine and Offsprings Health, Center for Global HealthNanjing Medical UniversityNanjing211166China
- School of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Leming Zheng
- State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, The Institute of Cardiovascular Sciences and Institute of Systems BiomedicinePeking UniversityBeijing100191China
| | - Peng Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
26
|
Das S, Lavine KJ. Role of Trained Immunity in Heath and Disease. Curr Cardiol Rep 2025; 27:18. [PMID: 39804563 DOI: 10.1007/s11886-024-02167-7] [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] [Accepted: 11/15/2024] [Indexed: 05/02/2025]
Abstract
PURPOSE OF REVIEW This review aims to explore the role of immune memory and trained immunity, focusing on how innate immune cells like monocytes, macrophages, and natural killer cells undergo long-term epigenetic and metabolic rewiring. Specifically, it examines the mechanisms by which trained immunity, often triggered by infection or vaccination, could impact cardiac processes and contribute to both protective and pathological responses within the cardiovascular system. RECENT FINDINGS Recent research demonstrates that vaccination and infection not only activate immune responses in circulating monocytes and tissue macrophages but also affect immune progenitor cells within the bone marrow environment, conferring lasting protection against heterologous infections. These protective effects are attributed to epigenetic and metabolic reprogramming, which enable a heightened immune response upon subsequent encounters with pathogens. However, while trained immunity is beneficial in combating infections, it has been linked to exacerbated inflammation, which may increase susceptibility to cardiovascular diseases, including atherosclerosis and heart failure. Our review highlights the dual nature of trained immunity: while it offers protective advantages against infections, it also poses potential risks for cardiovascular health by promoting chronic inflammation. Understanding the molecular mechanisms underlying immune memory's impact on cardiac processes could lead to new therapeutic strategies to mitigate cardiovascular diseases, such as atherosclerosis, heart failure, and diabetes. These insights build the grounds for future research to balance the benefits of trained immunity with its potential risks in cardiovascular disease management.
Collapse
Affiliation(s)
- Shibali Das
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8086, St. Louis, MO, 63110, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8086, St. Louis, MO, 63110, USA.
| |
Collapse
|
27
|
Fang H, Rodrigues e-Lacerda R, Barra NG, Kukje Zada D, Robin N, Mehra A, Schertzer JD. Postbiotic Impact on Host Metabolism and Immunity Provides Therapeutic Potential in Metabolic Disease. Endocr Rev 2025; 46:60-79. [PMID: 39235984 PMCID: PMC11720174 DOI: 10.1210/endrev/bnae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/18/2024] [Accepted: 09/04/2024] [Indexed: 09/07/2024]
Abstract
The gut microbiota influences aspects of metabolic disease, including tissue inflammation, adiposity, blood glucose, insulin, and endocrine control of metabolism. Prebiotics or probiotics are often sought to combat metabolic disease. However, prebiotics lack specificity and can have deleterious bacterial community effects. Probiotics require live bacteria to find a colonization niche sufficient to influence host immunity or metabolism. Postbiotics encompass bacterial-derived components and molecules, which are well-positioned to alter host immunometabolism without relying on colonization efficiency or causing widespread effects on the existing microbiota. Here, we summarize the potential for beneficial and detrimental effects of specific postbiotics related to metabolic disease and the underlying mechanisms of action. Bacterial cell wall components, such as lipopolysaccharides, muropeptides, lipoteichoic acids and flagellin, have context-dependent effects on host metabolism by engaging specific immune responses. Specific types of postbiotics within broad classes of compounds, such as lipopolysaccharides and muropeptides, can have opposing effects on endocrine control of host metabolism, where certain postbiotics are insulin sensitizers and others promote insulin resistance. Bacterial metabolites, such as short-chain fatty acids, bile acids, lactate, glycerol, succinate, ethanolamine, and ethanol, can be substrates for host metabolism. Postbiotics can fuel host metabolic pathways directly or influence endocrine control of metabolism through immunomodulation or mimicking host-derived hormones. The interaction of postbiotics in the host-microbe relationship should be considered during metabolic inflammation and metabolic disease.
Collapse
Affiliation(s)
- Han Fang
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Rodrigo Rodrigues e-Lacerda
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Dana Kukje Zada
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Nazli Robin
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Alina Mehra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| |
Collapse
|
28
|
Prajapati SK, Wang S, Mishra SP, Jain S, Yadav H. Protection of Alzheimer's disease progression by a human-origin probiotics cocktail. Sci Rep 2025; 15:1589. [PMID: 39794404 PMCID: PMC11724051 DOI: 10.1038/s41598-024-84780-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Microbiome abnormalities (dysbiosis) significantly contribute to the progression of Alzheimer's disease (AD). However, the therapeutic efficacy of microbiome modulators in protecting against these ailments remains poorly studied. Herein, we tested a cocktail of unique probiotics, including 5 Lactobacillus and 5 Enterococcus strains isolated from infant gut with proven microbiome modulating capabilities. We aimed to determine the probiotics cocktail's efficacy in ameliorating AD pathology in a humanized AD mouse model of APP/PS1 strains. Remarkably, feeding mice with 1 × 1011 CFU per day in drinking water for 16 weeks significantly reduced cognitive decline (measured by the Morris Water Maze test) and AD pathology markers, such as Aβ aggregation, microglia activation, neuroinflammation, and preserved blood-brain barrier (BBB) tight junctions. The beneficial effects were linked to a reduced inflammatory microbiome, leading to decreased gut permeability and inflammation in both systemic circulation and the brain. Although both male and female mice showed overall improvements in cognition and biological markers, females did not exhibit improvements in specific markers related to inflammation and barrier permeability, suggesting that the underlying mechanisms may differ depending on sex. In conclusion, our results suggest that this unique probiotics cocktail could serve as a prophylactic agent to reduce the progression of cognitive decline and AD pathology. This is achieved by beneficially modulating the microbiome, improving intestinal tight junction proteins, reducing permeability in both gut and BBB, and decreasing inflammation in the gut, blood circulation, and brain, ultimately mitigating AD pathology and cognitive decline.
Collapse
Affiliation(s)
- Santosh Kumar Prajapati
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shaohua Wang
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Biomedical Sciences, Infectious and Tropical Disease Institute, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Sidharth P Mishra
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA.
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Department of Internal Medicine-Digestive Diseases and Nutrition, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
29
|
Yadav H, Badal VD, Masternak MM, Leeuwenburgh C. Editorial for the special issue on "microbiomes in extremes of aging". Exp Gerontol 2025; 199:112663. [PMID: 39706502 DOI: 10.1016/j.exger.2024.112663] [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: 12/23/2024]
Abstract
This special issue of the Journal of Experimental Gerontology explores the dynamic interplay between microbiomes and aging-related conditions. The four selected studies highlight the role of microbiota in Alzheimer's disease, cancer immunotherapy, myocardial infarction and tryptophan metabolism, providing insights into how microbiomes influence health and disease in aging. These studies underscore the potential for microbiome-targeted interventions to mitigate aging-related disorders and improve the quality of life for older adults.
Collapse
Affiliation(s)
- Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| | - Varsha D Badal
- Department of Psychiatry, Stein Institute for Research on Aging, University of California San Diego, San Diego, CA, United States.
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States; Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland.
| | | |
Collapse
|
30
|
Soták M, Clark M, Suur BE, Börgeson E. Inflammation and resolution in obesity. Nat Rev Endocrinol 2025; 21:45-61. [PMID: 39448830 DOI: 10.1038/s41574-024-01047-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/30/2024] [Indexed: 10/26/2024]
Abstract
Inflammation is an essential physiological defence mechanism, but prolonged or excessive inflammation can cause disease. Indeed, unresolved systemic and adipose tissue inflammation drives obesity-related cardiovascular disease and type 2 diabetes mellitus. Drugs targeting pro-inflammatory cytokine pathways or inflammasome activation have been approved for clinical use for the past two decades. However, potentially serious adverse effects, such as drug-induced weight gain and increased susceptibility to infections, prevented their wider clinical implementation. Furthermore, these drugs do not modulate the resolution phase of inflammation. This phase is an active process orchestrated by specialized pro-resolving mediators, such as lipoxins, and other endogenous resolution mechanisms. Pro-resolving mediators mitigate inflammation and development of obesity-related disease, for instance, alleviating insulin resistance and atherosclerosis in experimental disease models, so mechanisms to modulate their activity are, therefore, of great therapeutic interest. Here, we review current clinical attempts to either target pro-inflammatory mediators (IL-1β, NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome, tumour necrosis factor (TNF) and IL-6) or utilize endogenous resolution pathways to reduce obesity-related inflammation and improve cardiometabolic outcomes. A remaining challenge in the field is to establish more precise biomarkers that can differentiate between acute and chronic inflammation and to assess the functionality of individual leukocyte populations. Such advancements would improve the monitoring of drug effects and support personalized treatment strategies that battle obesity-related inflammation and cardiometabolic disease.
Collapse
Affiliation(s)
- Matúš Soták
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Madison Clark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Bianca E Suur
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Emma Börgeson
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
31
|
Timmis K, Karahan ZC, Ramos JL, Koren O, Pérez‐Cobas AE, Steward K, de Lorenzo V, Caselli E, Douglas M, Schwab C, Rivero V, Giraldo R, Garmendia J, Turner RJ, Perlmutter J, Borrero de Acuña JM, Nikel PI, Bonnet J, Sessitsch A, Timmis JK, Pruzzo C, Prieto MA, Isazadeh S, Huang WE, Clarke G, Ercolini D, Häggblom M. Microbes Saving Lives and Reducing Suffering. Microb Biotechnol 2025; 18:e70068. [PMID: 39844583 PMCID: PMC11754571 DOI: 10.1111/1751-7915.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/24/2025] Open
Affiliation(s)
- Kenneth Timmis
- Institute of MicrobiologyTechnical University BraunschweigBraunschweigGermany
| | - Zeynep Ceren Karahan
- Department of Medical Microbiology and Ibn‐i Sina Hospital Central Microbiology LaboratoryAnkara University School of MedicineAnkaraTurkey
| | - Juan Luis Ramos
- Consejo Superior de Investigaciones Científicas, Estación Experimental del ZaidínGranadaSpain
| | - Omry Koren
- Azrieli Faculty of MedicineBar‐Ilan UniversitySafedIsrael
| | - Ana Elena Pérez‐Cobas
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS)Ramón y Cajal University HospitalMadridSpain
- CIBER in Infectious Diseases (CIBERINFEC)MadridSpain
| | | | - Victor de Lorenzo
- Department of Systems BiologyNational Centre of Biotechnology CSICMadridSpain
| | - Elisabetta Caselli
- Section of Microbiology, Department of Environmental and Prevention SciencesUniversity of FerraraFerraraItaly
| | - Margaret Douglas
- Usher InstituteUniversity of Edinburgh Medical School, and Public Health ScotlandEdinburghUK
| | - Clarissa Schwab
- Department of Biological and Chemical EngineeringAarhus UniversityAarhusDenmark
| | - Virginia Rivero
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Rafael Giraldo
- Department of Microbial BiotechnologyNational Centre for Biotechnology (CNB‐CSIC)MadridSpain
| | - Junkal Garmendia
- Instituto de AgrobiotecnologíaConsejo Superior de Investigaciones Científicas (IdAB‐CSIC)‐Gobierno de Navarra, MutilvaMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES)MadridSpain
| | - Raymond J. Turner
- Department of Biological SciencesUniversity of CalgaryCalgaryAlbertaCanada
| | | | | | - Pablo Ivan Nikel
- The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkLyngbyDenmark
| | - Jerome Bonnet
- Centre de Biochimie Structurale, INSERM/CNRSUniversity of MontpellierMontpellierFrance
| | - Angela Sessitsch
- Bioresources UnitAIT Austrian Institute of TechnologyViennaAustria
| | - James K. Timmis
- Department of Political ScienceUniversity of FreiburgFreiburgGermany
- Athena Institute for Research on Innovation and Communication in Health and Life SciencesVrije UniversiteitAmsterdamThe Netherlands
| | - Carla Pruzzo
- Department of Earth, Environmental and Life Sciences (DISTAV)University of GenoaGenovaItaly
| | - M. Auxiliadora Prieto
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Siavash Isazadeh
- Corporate Technical & PerformanceVeolia North AmericaParamusNew JerseyUSA
| | - Wei E. Huang
- Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Gerard Clarke
- APC Microbiome IrelandUniversity College CorkCorkIreland
- Department of Psychiatry & Neurobehavioral SciencesUniversity College CorkCorkIreland
| | - Danilo Ercolini
- Department of Agricultural SciencesUniversity of Naples Federico IINaplesItaly
| | - Max Häggblom
- Department of Biochemistry and Microbiology, RutgersThe State University of New JerseyNew BrunswickNew JerseyUSA
| |
Collapse
|
32
|
Hernandez J, Rodriguez JB, Trak-Fellermeier MA, Galvan R, Macchi A, Martinez-Motta P, Palacios C. Suboptimal vitamin D status and overweight/obesity are associated with gut integrity and inflammation in minority children and adolescents: A cross-sectional analysis from the MetA-bone trial. Nutr Res 2025; 133:13-21. [PMID: 39662375 DOI: 10.1016/j.nutres.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024]
Abstract
Preserving gut integrity is essential to preventing the development of chronic diseases. Several factors are associated with gut integrity and inflammation in adults. However, there is limited evidence in healthy children. This study evaluated the factors associated with gut integrity and inflammation in healthy children participating in the MetA-Bone trial. We hypothesized that age, sex, race/ethnicity, diet, vitamin D, and body composition will be associated with gut integrity and inflammation. Socio-demographic variables were collected with a questionnaire. Measures included markers of gut integrity (Intestinal Fatty Acid Binding Protein; I-FABP), and inflammation (IL-17 and calprotectin) determined by ELISA in 24-h urine and serum; serum 25(OH)D concentration (commercial lab), BMI percentile, and diet (24-h recalls). Analyses included descriptive statistics, chi-square, and adjusted logistic regressions. Participants (n=138) median age was 12.4 (11.1-13.3), 53.6% were male, 9.4% were Black/African American, and 71.1% were Hispanic/Latino. Children with suboptimal vitamin D were 3.35 times more likely to present gut integrity damage (elevated I-FABP) than those with optimal status (P = .005). Overweight/obesity and fructose intake were associated with inflammation (elevated calprotectin) (P < .05). Those with lower gut integrity damage had lower odds of having higher inflammation (P = .021). Other factors were not associated with inflammation. Suboptimal vitamin D status, overweight/obesity and inflammation may compromise the gut integrity in healthy children, suggesting an impairment on the intestinal barrier repair system. More research with a longitudinal design is needed to gain a deeper understanding of the role of additional factors linked to gut integrity and inflammation in healthy children.
Collapse
Affiliation(s)
- Jacqueline Hernandez
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA.
| | - Jose Bastida Rodriguez
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Maria Angelica Trak-Fellermeier
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Rodolfo Galvan
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Alison Macchi
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Preciosa Martinez-Motta
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Cristina Palacios
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| |
Collapse
|
33
|
Han X, Song Y, Piao Y, Wang Z, Li Y, Cui Q, Piao H, Yan G. Mechanism of miR-130b-3p in relieving airway inflammation in asthma through HMGB1-TLR4-DRP1 axis. Cell Mol Life Sci 2024; 82:9. [PMID: 39704848 DOI: 10.1007/s00018-024-05529-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024]
Abstract
Asthma is a chronic inflammatory respiratory disease characterized by recurrent breathing difficulties caused by airway obstruction and hypersensitivity. Although there is diversity in their specific mechanisms, microRNAs (miRNAs) have a significant impact on the development of asthma. Currently, the contribution of miR-130b-3p to asthma remains elusive. The goal of this study was to examine whether miR-130b-3p attenuates house dust mite (HDM)-induced asthma through High-mobility group box protein 1 (HMGB1)/Toll-like receptor 4 (TLR4)/mitochondrial fission protein (DRP1) signaling pathway. We elucidate that miR-130b-3p can bind to the HMGB1 3'UTR, attenuating HMGB1 mRNA and protein levels, and nucleo-cytoplasmic translocation of HMGB1. We observed that miR-130b-3p agomir or HMGB1 CKO attenuated HDM-induced airway inflammation and hyperresponsiveness, and decreased Th2-type cytokines in bronchoalveolar lavage fluid (BALF) and mediastinal lymph nodes. Further, HMGB1 CKO contributes to alleviating Th2 inflammation in AT-II cells (CD45.2-/CD31-/Epcam-+/proSP-C+/MHC-II+) from lung single cell suspensions of asthmatic mice by flow cytometry. Our findings identified miR-130b-3p as a potent regulator in asthma that exerts its anti-inflammatory effects by targeting HMGB1 and the subsequent HMGB1/TLR4/DRP1axis, presenting a prospective novel therapeutic avenue for asthma management.
Collapse
Affiliation(s)
- Xue Han
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, People's Republic of China
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, 133000, People's Republic of China
| | - Yilan Song
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, People's Republic of China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, No. 977, Gongyuan Road, Yanji, 133002, People's Republic of China
| | - Yihua Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, People's Republic of China
- Department of Emergency, Affiliated Hospital of Yanbian University, Yanji, 133000, People's Republic of China
| | - Zhiguang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, People's Republic of China
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, 133000, People's Republic of China
| | - Yan Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, People's Republic of China
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, 133000, People's Republic of China
| | - Qingsong Cui
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, People's Republic of China
- Department of Emergency, Affiliated Hospital of Yanbian University, Yanji, 133000, People's Republic of China
| | - Hongmei Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, People's Republic of China.
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, 133000, People's Republic of China.
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, People's Republic of China.
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, No. 977, Gongyuan Road, Yanji, 133002, People's Republic of China.
| |
Collapse
|
34
|
Wu T, Xu G, Hong X, Fan H, Zeng J, Liu Y, Hu J, Liang F, Yang J, Chen J. Acupuncture for hormonal readiness and gut microbiota in obese polycystic ovary syndrome: an open-label, randomized controlled trial. Front Endocrinol (Lausanne) 2024; 15:1509152. [PMID: 39749020 PMCID: PMC11693447 DOI: 10.3389/fendo.2024.1509152] [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: 10/10/2024] [Accepted: 11/20/2024] [Indexed: 01/04/2025] Open
Abstract
Objective To explore whether acupuncture combined with clomiphene can reduce the luteinizing hormone-to follicle-stimulating hormone ratio and impact the gut microbiota in patients with obese polycystic ovary syndrome. Methods This open-label, randomized, parallel-group controlled trial included 86 women aged 20-40 years with obese polycystic ovary syndrome and 19 healthy controls. Participants were randomly assigned to either an acupuncture combined with clomiphene group or a clomiphene-only group, with a healthy control group for comparison. The treatment lasted three menstrual cycles, with acupuncture administered three times weekly and clomiphene given daily from day five of menstruation for five consecutive days per cycle. The primary outcome was the change in the luteinizing hormone-to-follicle-stimulating hormone (LH/FSH) ratio. Secondary outcomes included levels of other sex hormones, glucose and lipid metabolism parameters, self-rating anxiety and depression scale scores, and gut microbiota composition. Results Intention-to-treat analysis showed that the adjusted mean luteinizing hormone-to follicle-stimulating hormone ratio decrease was -0.8 (95% CI: -1.14 to -0.46) in the acupuncture combined with clomiphene group and -0.22 (95% CI: -0.47 to 0.01) in the clomiphene group. The adjusted between-group difference was 0.53 (95% CI: 0.24 to 0.82, p < 0.001). The levels of Agathobacter faecis increased, and those of Erysipelatoclostridium spiroforme, Streptococcus lutetiensis, and Lactococcus lactis decreased after acupuncture combined with clomiphene treatment (p < 0.05). Conclusion Acupuncture combined with clomiphene may be safe and effective, reduce the luteinizing hormone-to follicle-stimulating hormone ratio, and improve insulin resistance in obese polycystic ovary syndrome, and these outcomes may be related to the gut microbiota.
Collapse
Affiliation(s)
- Tianyu Wu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Guixing Xu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine (Sichuan Hospital of Traditional Chinese Medicine), Chengdu, Sichuan, China
- Sichuan Clinical Research Center of Acupuncture and Moxibustion, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaojuan Hong
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Sichuan Clinical Research Center of Acupuncture and Moxibustion, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Huaying Fan
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jiuzhi Zeng
- Center of Reproductive Medicine, Sichuan Provincial Maternal and Child Health Hospital, Chengdu, Sichuan, China
| | - Yu Liu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jinqun Hu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Fanrong Liang
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Sichuan Clinical Research Center of Acupuncture and Moxibustion, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jie Yang
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Sichuan Clinical Research Center of Acupuncture and Moxibustion, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jiao Chen
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Sichuan Clinical Research Center of Acupuncture and Moxibustion, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
35
|
Gou Y, Lin F, Dan L, Zhang D. Exposure to toluene diisocyanate induces dysbiosis of gut-lung homeostasis: Involvement of gut microbiota. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125119. [PMID: 39414067 DOI: 10.1016/j.envpol.2024.125119] [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: 05/19/2024] [Revised: 09/29/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Toluene diisocyanate (TDI) is a major industrial compound that induces occupational asthma with steroid-resistant properties. Recent studies suggest that the gastrointestinal tract may be an effective target for the treatment of respiratory diseases. However, the alterations of the gut-lung axis in TDI-induced asthma remain unexplored. Therefore, in this study, a model of stable occupational asthma caused by TDI exposure was established to detect the alteration of the gut-lung axis. Exposure to TDI resulted in dysbiosis of the gut microbiome, with significant decreases in Barnesiella_intestinihominis, Faecalicoccus_pleomorphus, Lactobacillus_apodemi, and Lactobacillus_intestinalis, but increases in Alistipes_shahii and Odoribacter_laneus. The largest change in abundance was in Barnesiella_intestinihominis, which decreased from 12.14 per cent to 6.18 per cent. The histopathological abnormalities, including shorter length of intestinal villi, thinner thickness of muscularis, reduced number of goblet cells and inflammatory cell infiltration, were found in TDI-treated mice compared to control mice. In addition, increased permeability (evidenced by significantly reduced levels of ZO-1, Occludin and Claudin-1) and activation of TLR4/NF-κB signaling were observed in the intestine of these TDI-exposed mice. Concurrently, exposure to TDI resulted in airway hyperresponsiveness, overt cytokine production (e.g., IL-4, IL-5, IL-13, IL-25, and IL-33), and elevated IgE level within the respiratory tract. The expression of tight junction proteins is reduced and TLR4/NF-κB signaling is activated in the lung following TDI treatment. In addition, correlation analyses showed that changes in the gut microbiota were correlated with TDI exposure-induced airway inflammation. In conclusion, the present study suggests that the immune gut-lung axis may be involved in the development of TDI-induced asthma, which may have implications for potential interventions against steroid-resistant asthma.
Collapse
Affiliation(s)
- Yuxuan Gou
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China.
| | - Fu Lin
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| | - Li Dan
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| | - Dianyu Zhang
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| |
Collapse
|
36
|
Mignini I, Galasso L, Piccirilli G, Calvez V, Termite F, Esposto G, Borriello R, Miele L, Ainora ME, Gasbarrini A, Zocco MA. Interplay of Oxidative Stress, Gut Microbiota, and Nicotine in Metabolic-Associated Steatotic Liver Disease (MASLD). Antioxidants (Basel) 2024; 13:1532. [PMID: 39765860 PMCID: PMC11727446 DOI: 10.3390/antiox13121532] [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: 09/29/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025] Open
Abstract
Oxidative stress has been described as one of the main drivers of intracellular damage and metabolic disorders leading to metabolic syndrome, a major health problem worldwide. In particular, free radicals alter lipid metabolism and promote lipid accumulation in the liver, existing in the hepatic facet of metabolic syndrome, the metabolic dysfunction-associated steatotic liver disease (MASLD). Recent literature has highlighted how nicotine, especially if associated with a high-fat diet, exerts a negative effect on the induction and progression of MASLD by upregulating inflammation and increasing oxidative stress, abdominal fat lipolysis, and hepatic lipogenesis. Moreover, considerable evidence shows the central role of intestinal dysbiosis in the pathogenesis of MASLD and the impact of nicotine-induced oxidative stress on the gut microbiome. This results in an intricate network in which oxidative stress stands at the intersection point between gut microbiome, nicotine, and MASLD. The aim of this review is to delve into the molecular mechanisms linking tobacco smoking and MASLD, focusing on nicotine-induced microbiota modifications and their impact on MASLD development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Maria Assunta Zocco
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (I.M.); (L.G.); (G.P.); (V.C.); (F.T.); (G.E.); (R.B.); (L.M.); (M.E.A.); (A.G.)
| |
Collapse
|
37
|
Song RX, Ma XY, Zhou TT, Yu ZF, Wang J, Li BD, Jing YM, Wang H, Fu Y, Lv RZ, Jia SY, Li XM, Zhang LM. Excessive hydrogen sulfide-induced activation of NMDA receptors in the colon participates in anxiety- and compulsive-like behaviors in a rodent model of hemorrhagic shock and resuscitation. Int Immunopharmacol 2024; 142:113255. [PMID: 39332088 DOI: 10.1016/j.intimp.2024.113255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVE Hemorrhagic shock and resuscitation (HSR) cause inflammatory responses in the gastrointestinal tract and is associated with substantial morbidity and mortality rates. Hydrogen sulfide (H2S), a gasotransmitter with pleiotropic activity, exhibits anti-inflammatory benefits at physiological levels. However, deleterious effects are observed when its concentration increases. In this investigation, we employed a mouse model of HSR to examine the effects of an H2S scavenger on the gastrointestinal tract and brain, with emphasis on N-Methyl-d-Aspartate (NMDA) receptor function. METHODS Mice were immediately administered dl-propargylglycine (PAG) intragastrically as an H2S scavenger after HSR exposure. The O-maze and buried beads tests were used to assess compulsive- and anxiety-like behaviors. Pathological changes in the intestine were evaluated at 24 and 30 days after HSR. Subsequently, at 30 days after HSR, we examined electrophysiological and pathological changes in the amygdala. RESULTS Within 24 h of HSR exposure, animals treated with PAG showed significantly lower colonic injury. Additionally, compared to the HSR-treated mice 30 days after HSR, the PAG-treated mice displayed reduced buried beads, increased open-arm time, lower blood levels of Diamine Oxidase (DAO) and considerably improved ZO-1 intensity, a stronger association between the delta rhythm phase and beta activity amplitude, and lower neuroinflammatory response in the amygdala. MK-801, an NMDA receptor inhibitor, significantly reversed H2S-induced intestinal and cerebral injury. CONCLUSION This experimental data suggests that H2S-induced excessive activation of NMDA receptors contributes to anxiety- and compulsive-like behaviors caused by HSR.
Collapse
Affiliation(s)
- Rong-Xin Song
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No.2 Hospital), Cangzhou, China
| | - Xiao-Yi Ma
- Hebei University of Chinese Medicine, Shijiazhuang, China; Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China
| | - Ting-Ting Zhou
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China
| | - Zhi-Fang Yu
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China
| | - Jun Wang
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No.2 Hospital), Cangzhou, China
| | - Bao-Dong Li
- Department of Neurology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No.2 Hospital), Cangzhou, China
| | - Yu-Mo Jing
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No.2 Hospital), Cangzhou, China
| | - Han Wang
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No.2 Hospital), Cangzhou, China
| | - Yue Fu
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No.2 Hospital), Cangzhou, China
| | - Rui-Zhao Lv
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No.2 Hospital), Cangzhou, China
| | - Shi-Yan Jia
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China
| | - Xiao-Ming Li
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No.2 Hospital), Cangzhou, China; Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research, China.
| | - Li-Min Zhang
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No.2 Hospital), Cangzhou, China; Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China.
| |
Collapse
|
38
|
Upadhyay G, Gowda SGB, Mishra SP, Nath LR, James A, Kulkarni A, Srikant Y, Upendram R, Marimuthu M, Hui SP, Jain S, Vasundhara K, Yadav H, Halade GV. Targeted and untargeted lipidomics with integration of liver dynamics and microbiome after dietary reversal of obesogenic diet targeting inflammation-resolution signaling in aging mice. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159542. [PMID: 39097080 DOI: 10.1016/j.bbalip.2024.159542] [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: 12/20/2023] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Obesity, a global epidemic linked to around 4 million deaths yearly, arises from lifestyle imbalances impacting inflammation-related conditions like non-alcoholic fatty liver disease and gut dysbiosis. But the long-term effects of inflammation caused by lifestyle-related dietary changes remain unexplained. In this study, we used young male C57Bl/6 mice which were fed either an obesogenic diet (OBD) or a control diet (CON) for six months. Later, a group of mice from the OBD group were intervened to the CON diet (OBD-R) for four months, while another OBD group remained on the OBD diet. The OBD induced distinct changes in gut microbial, notably elevating Firmicutes and Actinobacteria, while reducing Bacteroidetes and Tenericutes. OBD-R restored microbial abundance like CON. Analyzing liver, plasma, and fecal samples revealed OBD-induced alterations in various structural and bioactive lipids, which were normalized to CON in the OBD-R, showcasing lipid metabolism flexibility and adaptability to dietary shifts. OBD increased omega 6 fatty acid, Arachidonic Acid (AA) and decreased omega 3-derived lipid mediators in the OBD mimicking non-alcoholic fatty liver disease thus impacting inflammation-resolution pathways. OBD also induced hepatic inflammation via increasing alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels and proinflammatory markers CCR2, TNF-α, and IL-1β in liver. Transitioning from OBD to CON mitigated inflammatory gene expression and restored lipid and cholesterol networks. This study underscores the intricate interplay between lifestyle-driven dietary changes, gut microbiota, lipid metabolism, and liver health. Notably, it suggests that shift from an OBD (omega-6 enriched) to CON partially alleviates signs of chronic inflammation during aging. Understanding these microbial, lipidomic, and hepatic inflammatory dynamics reveals potential therapeutic avenues for metabolic disorders induced by diet, emphasizing the pivotal role of diet in sustaining metabolic health.
Collapse
Affiliation(s)
- Gunjan Upadhyay
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Siddabasave Gowda B Gowda
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan; Graduate School of Global Food Resources, Hokkaido University, Sapporo, Japan
| | - Sidharth P Mishra
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA
| | - Lipsa Rani Nath
- Graduate School of Global Food Resources, Hokkaido University, Sapporo, Japan
| | - Adewale James
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA
| | - Alisha Kulkarni
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Yuktee Srikant
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Rohitram Upendram
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - MathanKumar Marimuthu
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA
| | - Kain Vasundhara
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA; Center for Aging and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Ganesh V Halade
- Heart Institute, Division of Cardiovascular Sciences, Department of Internal Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
39
|
Millman JF, Kondrashina A, Walsh C, Busca K, Karawugodage A, Park J, Sirisena S, Martin FP, Felice VD, Lane JA. Biotics as novel therapeutics in targeting signs of skin ageing via the gut-skin axis. Ageing Res Rev 2024; 102:102518. [PMID: 39389239 DOI: 10.1016/j.arr.2024.102518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/02/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024]
Abstract
Skin ageing is a phenomenon resulting from the aggregative changes to skin structure and function and is clinically manifested by physical features such as wrinkles, hyperpigmentation, elastosis, telangiectasia, and deterioration of skin barrier integrity. One of the main drivers of skin ageing, UV radiation, negatively influences the homeostasis of cells and tissues comprising the skin by triggering production of immune-mediated reactive oxygen species (ROS) and pro-inflammatory cytokines, as well as a various hormones and neuropeptides. Interestingly, an established link between the gut and the skin coined the 'gut-skin axis' has been demonstrated, with dysbiosis and gut barrier dysfunction frequently observed in certain inflammatory skin conditions and more recently, implicated in skin ageing. Therapeutic use of 'biotics' including prebiotics, probiotics, postbiotics, and synbiotics, which modulate the gut microbiota and production of microbially associated metabolites, influence the activity of the gut mucosal and immune systems and are showing promise as key candidates in addressing signs of skin ageing. In this review we aim to focus on the structure and function of the gut-skin axis and showcase the recent in-vitro and clinical evidence demonstrating the beneficial effects of select biotics in targeting signs of skin ageing and discuss the proposed mechanisms mediated via the gut-skin axis underpinning these effects.
Collapse
Affiliation(s)
- Jasmine F Millman
- Health and Happiness (H&H) Group, H&H Research, Level 6 & 7, 88 Langridge St, Collingwood, VIC 3066, Australia.
| | - Alina Kondrashina
- Health and Happiness (H&H) Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co., Cork P61K202, Ireland
| | - Clodagh Walsh
- Health and Happiness (H&H) Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co., Cork P61K202, Ireland
| | - Kizkitza Busca
- Health and Happiness (H&H) Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co., Cork P61K202, Ireland
| | - Aneesha Karawugodage
- Health and Happiness (H&H) Group, H&H Research, Level 6 & 7, 88 Langridge St, Collingwood, VIC 3066, Australia
| | - Julia Park
- Health and Happiness (H&H) Group, H&H Research, Level 6 & 7, 88 Langridge St, Collingwood, VIC 3066, Australia
| | - Sameera Sirisena
- Health and Happiness (H&H) Group, H&H Research, Level 6 & 7, 88 Langridge St, Collingwood, VIC 3066, Australia
| | - Francois-Pierre Martin
- Health and Happiness (H&H) Group, H&H Research, Avenue Sécheron 15 Bat F2/F3, Geneva 1202, Switzerland
| | - Valeria D Felice
- Health and Happiness (H&H) Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co., Cork P61K202, Ireland
| | - Jonathan A Lane
- Health and Happiness (H&H) Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co., Cork P61K202, Ireland.
| |
Collapse
|
40
|
Shao L, Zhang B, Song Y, Lyu Z, Zhang W, Yang W, Fu J, Li J, Shi J. Mucosal Hub Bacteria as Potential Targets for Improving High-Fat Diet-Related Intestinal Barrier Injury. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:3652740. [PMID: 39634324 PMCID: PMC11617042 DOI: 10.1155/cjid/3652740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
Background: Intestinal barrier injury contributes to multiple diseases such as obesity and diabetes, whereas no treatment options are available. Methods: Due to close interactions between mucosal microbiome and intestinal barrier, we evaluated the potential of mucosal bacteria in providing targets for high-fat diet (HFD)-related intestinal barrier injury. Whole-genome metagenomics was used to evaluate mucosal microbiome, while intestinal barrier injury was estimated using serum LPS, FITC-dextran intensity, and ZO-1 protein. Results: We found that HFD induced significant fat accumulation in epididymal tissue at weeks 4 and 12, while ALT, LDL, and TC increased at week 12. Intestinal barrier injury was confirmed by elevated serum LPS at both weeks, upregulated FITC-dextran intensity, and decreased ZO-1 protein at week 12. Fourteen species such as Phocaeicola vulgatus differed in HFD-fed mice. The co-occurrence network of mucosal microbiome shifted from scale-free graph in controls to nearly random graph in HFD-fed mice. Besides, 10 hub bacteria especially Bacteroides ovatus decreased drastically in both mucosal and fecal samples of HFD-fed mice, correlated with intestinal permeability, ALT, and KEGG pathways such as "Mitochondrial biogenesis" and "metabolism". Moreover, Bacteroides ovatus has been confirmed to improve intestinal barrier function in a recent study. Conclusion: Mucosal hub bacteria can provide potential targets for improving HFD-related intestinal barrier function.
Collapse
Affiliation(s)
- Li Shao
- School of Clinical Medicine, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
| | - Binbin Zhang
- School of Clinical Medicine, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
| | - Yu Song
- Department of Hepatology, 2nd Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Zhe Lyu
- Department of Stomatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China
| | - Weishi Zhang
- Department of Otolaryngology, Affiliated Hospital 2 of Nantong University, Nantong 226001, Jiangsu, China
| | - Wenjun Yang
- School of Clinical Medicine, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China
| | - Jinlong Fu
- School of Clinical Medicine, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang, China
| | - Jie Li
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Junping Shi
- School of Clinical Medicine, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China
- Department of Hepatology, 2nd Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| |
Collapse
|
41
|
Smolińska K, Hułas-Stasiak M, Dobrowolska K, Sobczyński J, Szopa A, Tomaszewska E, Muszyński S, Smoliński K, Dobrowolski P. Effects of an Innovative High-Fat Diet on Intestinal Structure, Barrier Integrity, and Inflammation in a Zebrafish Model of Visceral Obesity. Int J Mol Sci 2024; 25:12723. [PMID: 39684433 DOI: 10.3390/ijms252312723] [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: 10/20/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
High-fat diet (HFD)-induced obesity is a global health concern associated with gastrointestinal disorders. While mammalian models have elucidated the effects of a HFD on intestinal structure and function, its impact on zebrafish, a crucial model for studying diet-induced obesity and gastrointestinal dysfunction, remains inadequately characterized. This study investigated the influence of a HFD on zebrafish intestinal morphology, tight junction (TJ) protein expression, and inflammatory markers. Zebrafish fed a control diet or HFD with 40% or 60% fat exhibited significant alterations in intestinal morphology, with increased villi number but reduced villi width and length, suggesting compensatory responses to dietary stress. TJ protein expression (Claudin 2, Claudin 3, and Claudin 10) showed complex changes, particularly in the HFD60 juvenile group, indicating a multifaceted response in barrier integrity. Pro-inflammatory cytokine IL-6 and TNF-α levels were lower in both the juvenile and adult HFD60 groups than in the HFD40 and control groups, while elevated anti-inflammatory IL-10 levels in HFD60 adult zebrafish suggested activation of compensatory mechanisms. These findings highlight zebrafish as a valuable model for studying the effects of HFD on intestinal health and provide insights into the relationship between dietary fat, gut dysfunction, and inflammation.
Collapse
Affiliation(s)
- Katarzyna Smolińska
- Chronic Wounds Laboratory, Medical University of Lublin, Chodźki St. 7, 20-093 Lublin, Poland
| | - Monika Hułas-Stasiak
- Department of Functional Anatomy and Cytobiology, Maria Curie Sklodowska University, Akademicka St. 19, 20-033 Lublin, Poland
| | - Katarzyna Dobrowolska
- Faculty of Biology and Biotechnology, Maria Curie Sklodowska University, Akademicka St. 19, 20-033 Lublin, Poland
| | - Jan Sobczyński
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, Chodźki St. 7, 20-093 Lublin, Poland
| | - Aleksandra Szopa
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, Chodźki St. 7, 20-093 Lublin, Poland
| | - Ewa Tomaszewska
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka St. 12, 20-950 Lublin, Poland
| | - Siemowit Muszyński
- Department of Biophysics, University of Life Sciences in Lublin, Akademicka St. 13, 20-950 Lublin, Poland
| | - Kacper Smoliński
- Faculty of Biology, Warsaw University, Żwirki i Wigury St. 101, 02-089 Warsaw, Poland
| | - Piotr Dobrowolski
- Department of Functional Anatomy and Cytobiology, Maria Curie Sklodowska University, Akademicka St. 19, 20-033 Lublin, Poland
| |
Collapse
|
42
|
Wang K, Cunha E Rocha K, Qin H, Zeng Z, Ying W. Host metabolic inflammation fueled by bacterial DNA. Trends Endocrinol Metab 2024:S1043-2760(24)00294-7. [PMID: 39609222 DOI: 10.1016/j.tem.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 11/30/2024]
Abstract
Metabolic diseases, characterized by chronic low-grade inflammation, exhibit a compromised gut barrier allowing the translocation of bacteria-derived products to bloodstream and distant metabolic organs. Bacterial DNA can be detected in metabolic tissues during the onset of these diseases, highlighting its role in the development of metabolic diseases. Extracellular vesicles (EVs) are involved in the delivery of bacterial DNA to the local tissues, and its sensing by the host triggers local and system inflammation. Understanding bacterial DNA translocation and its induced inflammation is crucial in deciphering metabolic disease pathways. Here, we delve into the mechanisms dictating the interaction between host physiology and bacterial DNA, focusing on its origin and delivery, host immune responses against it, and its roles in metabolic disorders.
Collapse
Affiliation(s)
- Ke Wang
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Karina Cunha E Rocha
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Houji Qin
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Zixuan Zeng
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
43
|
Protasiewicz-Timofticiuc DC, Bădescu D, Moța M, Ștefan AG, Mitrea A, Clenciu D, Efrem IC, Roșu MM, Vladu BE, Gheonea TC, Moța E, Vladu IM. Back to Roots: Dysbiosis, Obesity, Metabolic Syndrome, Type 2 Diabetes Mellitus, and Obstructive Sleep Apnea-Is There an Objective Connection? A Narrative Review. Nutrients 2024; 16:4057. [PMID: 39683451 DOI: 10.3390/nu16234057] [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: 10/27/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
In recent decades, it has become clear that the gut is more than just a digestive organ; it also functions as an immune organ with regulatory capabilities and acts as a "second brain" that influences brain function due to the presence and regulatory roles of the gut microbiota (GM). The GM is a crucial component of its host and significantly impacts human health. Dysbiosis, or microbial imbalance, has been closely linked to various diseases, including gastrointestinal, neurological, psychiatric, and metabolic disorders. The aim of this narrative review is to highlight the roles of the GM in maintaining metabolic health. Sleep is a vital biological necessity, with living organisms having evolved an internal sleep-wake rhythm that aligns with a roughly 24 h light/dark cycle, and this is known as the circadian rhythm. This cycle is essential for tissue repair, restoration, and overall optimal body functioning. Sleep irregularities have become more prevalent in modern society, with fast-paced lifestyles often disrupting normal sleep patterns. Urban living factors, such as fast food consumption, shift work, exposure to artificial light and nighttime noise, medications, and social activities, can adversely affect circadian rhythms, with dysbiosis being one of the many factors incriminated in the etiology of sleep disorders.
Collapse
Affiliation(s)
| | - Diana Bădescu
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Maria Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | | | - Adina Mitrea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Clenciu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ion Cristian Efrem
- Department of Medical Semiology, Faculty of Dentistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Maria Magdalena Roșu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Beatrice Elena Vladu
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Theodora Claudia Gheonea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Eugen Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ionela Mihaela Vladu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
44
|
Li W, Xia Y, Yang J, Sanyal AJ, Shah VH, Chalasani NP, Yu Q. Disrupted balance between pro-inflammatory lipid mediators and anti-inflammatory specialized pro-resolving mediators is linked to hyperinflammation in patients with alcoholic hepatitis. Front Immunol 2024; 15:1377236. [PMID: 39640267 PMCID: PMC11617321 DOI: 10.3389/fimmu.2024.1377236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Background Alcoholic hepatitis (AH) is characterized by intense systemic and liver inflammation, posing significant risks of health complications and mortality. While inflammation is a crucial defense mechanism against injury and infection, its timely resolution is essential to prevent tissue damage and restore tissue homeostasis. The resolution of inflammation is primarily governed by specialized pro-resolving mediators (SPMs), lipid metabolites derived from w-6 and w-3 poly-unsaturated fatty acids (PUFAs). Currently, the balance between pro-inflammatory lipid mediators (PLMs) and SPMs in the w-6 and w-3 PUFA metabolic pathways and the impact of alcohol abstinence on profiles of PLMs and SPMs in AH patients are not well studied. Methods In this study, we used LC-MS/MS and ELISA to quantify levels of lipid mediators (LMs) and their precursors in the plasma samples from 58 AH patients, 29 heavy drinkers without overt liver diseases (HDCs), and 35 healthy controls (HCs). Subsequently, we assessed correlations of altered LMs with clinical parameters and inflammatory mediators. Furthermore, we conducted a longitudinal study to analyze the effects of alcohol abstinence on LMs over 6- and 12-month follow-ups. Results AH patients exhibited significantly higher plasma levels of w-6 PLMs (PGD2 and LTB4) and SPM RvE1 compared to HDCs or HCs. Conversely, the SPM LXA4 was significantly downregulated in AH patients. Some of these altered LMs were found to correlate with AH disease severity and various inflammatory cytokines. Particularly, the LTB4/LXA4 ratio was substantially elevated in AH patients relative to HDCs and HCs. This altered ratio displayed a positive correlation with the MELD score. Importantly, the majority of dysregulated LMs, particularly PLMs, were normalized following alcohol abstinence.
Collapse
Affiliation(s)
- Wei Li
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ying Xia
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jing Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Arun J. Sanyal
- Division of Gastroenterology and Hepatology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Naga P. Chalasani
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Qigui Yu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
45
|
Rust BM, Nielsen FH, Yan L. Dietary Intake of Chromista Oil Alters Hepatic Metabolomic Profile of Mice With Excess Fat Mass. Nutr Metab Insights 2024; 17:11786388241297143. [PMID: 39568657 PMCID: PMC11577470 DOI: 10.1177/11786388241297143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024] Open
Abstract
Increasing dietary intake of fish oil is frequently recommended for decreasing the risk for cardiovascular diseases and improving metabolic health. We hypothesised that dietary intake of chromista oil (a marine food product and a rich source of long-chain n-3 polyunsaturated fatty acids) ameliorates metabolic impairments in mice with established excess adiposity. Three-to 4-week-old mice (male) were fed a control (n = 12) or a high-fat diet (HFD, n = 24) for 12 weeks to establish body fat mass. Then, mice on the HFD were assigned to 2 groups (n = 12 each) with 1 continuing being fed the HFD and the other fed the HFD with chromista oil for an additional 12 weeks. Intake of chromista oil did not affect body weight and body adiposity of the mice fed the HFD; mice fed the HFD had significantly more body weight and fat mass than control mice. The flattened daily oscillations of respiratory exchange ratio induced by the HFD were not changed by chromista oil intake. Intake of chromista oil significantly increased plasma concentration of insulin, the calculated value of HOMA-IR, and plasma concentration of adiponectin in the mice fed the HFD. However, blood glucose was unaffected by chromista oil. Transcription of genes encoding circadian rhythm and fatty acid metabolism of the 2 HFD-fed groups were similar. Untargeted metabolomic analysis showed that intake of chromista oil altered the hepatic metabolomic profile with substantial alterations in amino acid metabolism. Findings from this study indicate that dietary intake of chromista oil does not improve glucose homeostasis or alter the diminished metabolic flexibility in mice with excess adiposity induced by the HFD. argeted metabolomic analysis is warranted to investigate the effects of dietary chromista oil, as a source of n-3 poly unsaturated fatty acids, on metabolism in models of obesity.
Collapse
Affiliation(s)
- Bret M Rust
- U.S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND, USA
| | - Forrest H Nielsen
- U.S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND, USA
| | - Lin Yan
- U.S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND, USA
| |
Collapse
|
46
|
Lin L, Xiang S, Chen Y, Liu Y, Shen D, Yu X, Wu Z, Sun Y, Chen K, Luo J, Wei G, Wang Z, Ning Z. Gut microbiota: Implications in pathogenesis and therapy to cardiovascular disease (Review). Exp Ther Med 2024; 28:427. [PMID: 39301250 PMCID: PMC11411594 DOI: 10.3892/etm.2024.12716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/03/2024] [Indexed: 09/22/2024] Open
Abstract
The gut microbiota refers to the diverse bacterial community residing in the gastrointestinal tract. Recent data indicate a strong correlation between alterations in the gut microbiota composition and the onset of various diseases, notably cardiovascular disorders. Evidence suggests the gut-cardiovascular axis signaling molecules released by the gut microbiota play a pivotal role in regulation. This review systematically delineates the association between dysbiosis of the gut microbiota and prevalent cardiovascular diseases, including atherosclerosis, hypertension, myocardial infarction and heart failure. Furthermore, it provides an overview of the putative pathogenic mechanisms by which dysbiosis in the gut microbiota contributes to the progression of cardiovascular ailments. The potential modulation of gut microbiota as a preventive strategy against cardiovascular diseases through dietary interventions, antibiotic therapies and probiotic supplementation is also explored and discussed within the present study.
Collapse
Affiliation(s)
- Li Lin
- Department of Biochemistry, Basic Medicine School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Shaowei Xiang
- Department of Neurosurgery, Enshi State Central Hospital, Enshi, Hubei 445000, P.R. China
| | - Yuan Chen
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Yan Liu
- Department of Internal Medicine, The Second Affiliated Hospital, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Dingwen Shen
- Department of Parasitology, Basic Medicine School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Xiaoping Yu
- Department of Function, The Second Affiliated Hospital, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zhe Wu
- Department of Histology and Embryology, Basic Medicine School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Yanling Sun
- Department of Histology and Embryology, Basic Medicine School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Kequan Chen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Jia Luo
- School of Sport, Xianning Vocational and Technical College, Xianning, Hubei 437100, P.R. China
| | - Guilai Wei
- School of Art and Design, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zhiguo Wang
- Department of Dermatology, The First Affiliated Hospital, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zhifeng Ning
- Department of Human Anatomy, Basic Medicine School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|
47
|
Li Y, Sun M, Tian X, Bao T, Yu Q, Ma NL, Gan R, Cheang WS, Wu X. Gymnemic acid alleviates gut barrier disruption and lipid dysmetabolism via regulating gut microbiota in HFD hamsters. J Nutr Biochem 2024; 133:109709. [PMID: 39053860 DOI: 10.1016/j.jnutbio.2024.109709] [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: 01/23/2024] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Gut microbiota dysbiosis and gut barrier disruption are key events associated with high-fat diet (HFD)-induced systemic metabolic disorders. Gymnemic acid (GA) has been reported to have an important role in alleviating HFD-induced disorders of glycolipid metabolism, but its regulatory role in HFD-induced disorders of the gut microbiota and gut barrier function has not been elucidated. Here we showed that GA intervention in HFD-induced hamsters increased the relative abundance of short-chain fatty acid (SCFA)-producing microbes including Lactobacillus (P<.05) and Lachnoclostridium (P<.01) in the gut, and reduced the relative abundance of lipopolysaccharide (LPS)-producing microbes including Enterococcus (P<.05) and Bacteroides (P<.05), subsequently improving HFD-induced intestinal barrier dysfunction and systemic inflammation. Specifically, GA intervention reduced mRNA expression of inflammatory cytokines, including IL-1β, IL-6, and TNF-α (P<.01), increased mRNA expression of antioxidant-related genes, including Nfe2l2, Ho-1, and Nqo1 (P<.01), and increased mRNA expression of intestinal tight junction proteins, including Occludin and Claudin-1 (P<.01), thereby improving gut barrier function of HFD hamsters. This ameliorative effect of GA on the gut of HFD hamsters may further promote lipid metabolic balance in liver and adipose tissue by regulating the Toll-like receptor 4 (TLR4)-nuclear factor-κB (NF-κB) signaling pathway. Taken together, these results systematically revealed the important role of GA in regulating HFD-induced gut microbiota disturbance and gut barrier function impairment, providing a potential clinical theoretical basis for targeted treatment of HFD-induced microbiota dysbiosis.
Collapse
Affiliation(s)
- Yumeng Li
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin, China; TIB-UM Joint Laboratory of Synthetic Biology for Traditional Chinese Medicine, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, China
| | - Mingzhe Sun
- Air Force Medical Center of People's Liberation Army, Beijing, China; College of food science & nutritional engineering, China Agricultural University, Beijing, China
| | - Xutong Tian
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin, China
| | - Tongtong Bao
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin, China
| | - Qian Yu
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin, China
| | - Nyuk Ling Ma
- BIOSES Research Interest Group, Faculty of Science & Marine Environment, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia; Center for Global Health Research (CGHR), Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Renyou Gan
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore; Department of Food Science and Nutrition, Faculty of Science, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China; TIB-UM Joint Laboratory of Synthetic Biology for Traditional Chinese Medicine, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, China
| | - Xin Wu
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin, China; TIB-UM Joint Laboratory of Synthetic Biology for Traditional Chinese Medicine, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, China.
| |
Collapse
|
48
|
Oikonomou I, Papageorgiou A, de Lastic AL, Moulias A, Georgopoulou GA, Mouzaki A, Koufou EE, Tsigkas G, Gogos C, Davlouros P, Assimakopoulos SF. Gut barrier dysfunction, endotoxemia and inflammatory response in STEMI patients and effect of primary PCI. Am J Med Sci 2024; 368:485-493. [PMID: 38969287 DOI: 10.1016/j.amjms.2024.07.004] [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: 12/22/2023] [Revised: 06/05/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Gut-derived bacterial and endotoxin translocation induce systemic inflammation, which exerts a pivotal pathogenetic role in all phases of atherosclerosis. OBJECTIVES To investigate prospectively the gut barrier function, endotoxin translocation and inflammatory response in ST-elevation myocardial infarction (STEMI) patients undergoing primary percutaneous coronary artery intervention (PPCI). METHODS Twenty-seven patients with STEMI that underwent successful PPCI were subjected to peripheral blood sampling at 3-time points; before PPCI (day0), 24 h (day1) and 96 h (day4) after PPCI and were compared with 20 chronic coronary syndrome (CCS) patients and 11 healthy controls. Serum ZO-1, I-FABP and endotoxin concentrations were determined by ELISA. Concentrations of cytokines IL-1β, -6, -8, -10 and TNF-α were determined by flow cytometry. RESULTS Patients with STEMI before PPCI (day0) had increased serum ZO-1 and endotoxin, both at significantly higher levels compared to CCS patients. STEMI induced also significant increases of the cytokines IL-6, -8 and -10. After PPCI, a significant improvement of gut barrier integrity (ZO-1) and endotoxemia was observed from the first day. At day4 post PPCI, systemic endotoxin and cytokines IL-6, -8 and -10 levels were reduced to control levels. Serum ZO-1 levels were positively correlated with systemic IL-10 concentrations (r = 0.471). CONCLUSION STEMI is associated with gut barrier dysfunction, systemic endotoxemia and inflammatory response, which improve rapidly following successful PPCI.
Collapse
Affiliation(s)
- Ioanna Oikonomou
- Department of Internal Medicine and Division of Infectious Diseases, University of Patras Medical School, Patras, Greece
| | - Angeliki Papageorgiou
- Division of Cardiology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Anne-Lise de Lastic
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Athanasios Moulias
- Division of Cardiology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | | | - Athanasia Mouzaki
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Eleni-Evangelia Koufou
- Division of Cardiology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Grigorios Tsigkas
- Division of Cardiology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Charalambos Gogos
- Department of Internal Medicine and Division of Infectious Diseases, University of Patras Medical School, Patras, Greece
| | - Periklis Davlouros
- Division of Cardiology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Stelios F Assimakopoulos
- Department of Internal Medicine and Division of Infectious Diseases, University of Patras Medical School, Patras, Greece.
| |
Collapse
|
49
|
Hakkak R, Korourian S, Li W, Spray B, Twaddle NC, Randolph CE, Børsheim E, Robeson II MS. Dietary soy protein reverses obesity-induced liver steatosis and alters fecal microbial composition independent of isoflavone level. Front Nutr 2024; 11:1487859. [PMID: 39529929 PMCID: PMC11551038 DOI: 10.3389/fnut.2024.1487859] [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: 08/28/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major public health concern that is exacerbated by the obesity pandemic. Dietary interventions have the potential to alleviate obesity-associated MASLD through variable mechanisms, including optimizing the gut microbiota. Previously, we reported that soy protein concentrate (SPC) with low or high levels of isoflavone (LIF or HIF) protected young obese Zucker rats from developing liver steatosis. The current study was designed to test whether SPC-LIF and SPC-HIF diets would reverse liver steatosis and alter fecal microbial composition in adult obese Zucker rats with existing steatosis. Methods Six-week-old male obese Zucker rats (n = 26) were fed a casein control diet (CAS) for 8 weeks and 7 rats were randomly selected and sacrificed to confirm liver steatosis. The remaining rats were randomly assigned to receive CAS, SPC-LIF, or SPC-HIF diet (n = 6-7/group) for an additional 10 weeks. Results Compared to CAS diet, feeding SPC-LIF and SPC-HIF diets resulted in significantly lower liver weight, liver steatosis score, and liver microvesicular score (p < 0.05), but did not lead to difference in body weight, liver macrovesicular score, serum ALT, or serum AST. Isoflavone levels (e.g., LIF vs. HIF) did not affect any of these measurements except in the SPC-HIF group, which had an additional decrease in liver weight (p < 0.05) compared to the SPC-LIF group. The SPC-HIF group also had significantly higher levels of the aglycone forms of daidzein, genistein, and equol as well as the total levels of daidzein, genistein, and equol compared to SPC-LIF or CAS diet fed rats (p < 0.05). The distribution of microbial communities based on measures of beta diversity of both SPC-LIF and SPC-HIF groups were significantly different to that of the CAS group (p ≤ 0.005). Alpha-diversity did not differ between any of the groups. Conclusion Taken together, dietary soy protein can reverse liver steatosis in adult Zucker rats, and the reversal of steatosis is accompanied by alterations in gut microbial composition.
Collapse
Affiliation(s)
- Reza Hakkak
- Department of Dietetics and Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children’s Research Institute, Little Rock, AR, United States
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Soheila Korourian
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Wei Li
- Department of Dietetics and Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Beverly Spray
- Division of Biostatistics Core, Arkansas Children’s Research Institute, Little Rock, AR, United States
| | - Nathan C. Twaddle
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | | | - Elisabet Børsheim
- Arkansas Children’s Research Institute, Little Rock, AR, United States
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children’s Nutrition Center, Little Rock, AR, United States
| | - Michael S. Robeson II
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
50
|
Wang X, Guo Q, Liu Z, Wang Y, Cao C, Jin L, Li C, Xiao J, Zhao W. Alterations in the Gut Microbiota Composition in Obesity with and without Type 2 Diabetes: A Pilot Study. Diabetes Metab Syndr Obes 2024; 17:3965-3974. [PMID: 39469300 PMCID: PMC11514687 DOI: 10.2147/dmso.s477494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
Purpose Obesity has become a major public health concern worldwide, increasing the risk of T2DM. Growing evidence indicates gut microbiota dysbiosis is related to metabolic disorders. We aimed to firstly investigate the compositional and functional features of the gut microbiome between obesity with and without T2DM in the Chinese population. Methods A total of 32 obese individuals accompanied with T2DM and 18 age and gender-matched obesity with normal glucose tolerance (NGT) were enrolled. Fecal samples were collected, and the gut microbiota profile was determined using the Illumina MiSeq platform based on V3-V4 bacterial 16S rRNA gene. Results Compared with obesity- NGT, obesity-T2DM showed a significantly higher alpha diversity. Principal coordinates analysis based on both Bray-Curtis distance and weighted Unifrac revealed that the global microbial composition was significantly different between the two groups (P = 0.007 and P = 0.005, respectively). At the phylum level, Obesity-T2DM patients exhibited a significant decrease in Bacteroidetes, and a pronounced increase in Firmicutes. Regarding the genus level, Bacteroides and Escherichia-Shigella were found to increase considerably, while Prevotella_9 and Sutterella had an evident decrease in Obesity-T2DM. Furthermore, Spearman correlation analysis revealed that Prevotella_9 and Sutterella were negatively associated with HbA1c and fasting blood glucose. Conclusion We found clear differences in the gut microbiota composition in obesity-T2DM compared with obesity-NGT. Obesity accompanied with T2DM may aggravate the obesity-associated gut microbiota, and gut microbiota is expected to be a promising novel intervention target for obese management. However, larger sample size and more in-depth taxonomic identification studies are warranted.
Collapse
Affiliation(s)
- Xiaojing Wang
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Qinli Guo
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Zhaoxiang Liu
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Yanlei Wang
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Chenxiang Cao
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Lixia Jin
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Caihong Li
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Jianzhong Xiao
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Wenhui Zhao
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| |
Collapse
|