1
|
Tao Y, Zeng Y, Zeng R, Gou X, Zhou X, Zhang J, Nhamdriel T, Fan G. The total alkaloids of Berberidis Cortex alleviate type 2 diabetes mellitus by regulating gut microbiota, inflammation and liver gluconeogenesis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118957. [PMID: 39426578 DOI: 10.1016/j.jep.2024.118957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/24/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Type 2 diabetes mellitus (T2DM) has become a public health problem worldwide. There is growing interest in finding drugs to treat T2DM from herbal medicine. Berberidis Cortex is a traditional Tibetan herb commonly used in the treatment of T2DM, and alkaloids are its main active components. However, the anti-diabetic mechanisms of the total alkaloids of Berberidis Cortex (TBC) remain unclear. AIM OF THE STUDY The aim of this study was to evaluate the anti-T2DM efficacy of TBC and reveal the mechanisms behind its effects. MATERIALS AND METHODS UPLC-Q-Exactive Orbitrap MS technology was employed to qualitatively identify alkaloid components in TBC. T2DM rat models were induced by high-fat diet combined with streptozotocin, and then treated with different doses of TBC (43.5, 87, 174 mg/kg/d) for 40 days. Biochemical parameters, such as fasting blood glucose (FBG), oral glucose tolerance test (OGTT), glycated serum protein (GSP), homeostatic model assessment of insulin resistance (HOMA-IR), total cholesterol (TC), triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C), alongside H&E and PAS staining were used to evaluate the anti-diabetic activity of TBC. More importantly, metagenomics, transcriptomics, targeted metabolomics, and Western blot analysis were integrated to reveal the underlying mechanisms of TBC for T2DM treatment. RESULTS TBC significantly reduced the levels of FBG, OGTT, GSP, HOMA-IR, TC, TG, and LDL-C, and improved the histopathological alterations of pancreatic and liver tissues in T2D rats. It also reduced serum levels of lipopolysaccharide (LPS) and several pro-inflammatory cytokines (IL-6, IL-1β and TNF-α). Gut microbiome analysis by metagenomics proved that TBC could improve gut microbiota dysbiosis, including an increase in some beneficial bacteria (e.g., Bifidobacterium pseudolongum and Lactobacillus acidophilus) and a decrease in some harmful bacteria (e.g., Marvinbryantia and Parabacteroides). Western blot analysis found that TBC significantly up-regulated the expression of three intestinal barrier related tight junction proteins (ZO-1, occludin, and claudin-1), and effectively suppressed several key proteins in the TLR4/MyD88/NF-κB inflammatory cascade, including TLR4, MyD88 and p-NF-κB p65. Moreover, hepatic transcriptomics analysis further revealed the regulatory role of TBC on gluconeogenesis related genes, such as Pgc, and Creb1. Targeted metabolomics and Western blot analysis showed that TBC improved BAs dysregulation in T2DM rats, specifically increasing TCDCA and CA levels, thereby activating several proteins in the FXR/FGF15 signaling axis (i.e., FXR, FGF15 and FGFR4), and then decreased the expression of p-CREB1 and PGC-1α to inhibit liver gluconeogenesis. CONCLUSIONS TBC can significantly improve hyperglycemia, insulin resistance, hyperlipidemia, and inflammation in T2DM rats. The mechanism is related to the regulation of multiple links, including improving gut microbiota dysbiosis, protecting the intestinal barrier by up-regulating the expression of three tight junction proteins, reducing inflammation by inhibiting the LPS/TLR4/MyD88/NF-κB pathway, and inhibiting liver gluconeogenesis by regulating BAs/FXR/FGF15 and CREB1/PGC-1α signaling pathways.
Collapse
Affiliation(s)
- Yiwen Tao
- School of Ethnic Medicine, School of Pharmacy and Meishan Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yujiao Zeng
- School of Ethnic Medicine, School of Pharmacy and Meishan Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Rui Zeng
- School of Ethnic Medicine, School of Pharmacy and Meishan Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiaoling Gou
- School of Ethnic Medicine, School of Pharmacy and Meishan Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianhua Zhou
- School of Ethnic Medicine, School of Pharmacy and Meishan Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jing Zhang
- School of Ethnic Medicine, School of Pharmacy and Meishan Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Tsedien Nhamdriel
- Department of Tibetan Medicine, University of Tibetan Medicine, Lhasa, 850000, China.
| | - Gang Fan
- School of Ethnic Medicine, School of Pharmacy and Meishan Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
2
|
Yang Y, Miao J, Du J, Xu S, Zhang K, Wu T, Tao C, Wang Y, Fang M, Yang S. Deficiency of SLC26A3 promotes jejunal barrier damage in metabolic disease-susceptible transgenic pigs. Int J Biol Macromol 2024; 281:136245. [PMID: 39368571 DOI: 10.1016/j.ijbiomac.2024.136245] [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: 07/02/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Intestinal disorders are common in metabolic syndrome. However, their pathogenesis is still not fully understood. Pig and human intestines are highly similar in terms of associated metabolic processes. Here, we successfully constructed a metabolic disease-susceptible transgenic (TG) Bama pig model by knocking in three humanized disease risk genes with the CRISPR/Cas9 technique to assess its potential as a model for human intestinal diseases and explore the possible pathological mechanisms involved. We found that jejunal barrier integrity was disrupted and that the infiltration of inflammatory cells increased in TG pigs after high-fat and high-sucrose diet (HFHSD) treatment. We revealed significant differences in the transcriptome, associated microbiome profiles and microbial metabolite short-chain fatty acid (SCFA) content of the jejunum of TG pigs. Notably, we found that SLC26A3 was significantly downregulated in TG pigs. Knockdown or overexpression of the SLC26A3 gene in IPEC-J2 cells significantly affected the expression of MUC2, MUC13 and occludin. Furthermore, in vitro experiments further verified that CDX2 directly regulated the expression of SLC26A3. Mechanistically, CDX2 mediated intestinal barrier function by enhancing the expression of SLC26A3 by binding to its promoter region between -1120 and - 1070 bp. TG pigs represent a promising model that provides new insights into preclinical research on human intestinal metabolic diseases associated with metabolic disorders and revealed that SLC26A3 may be a potential therapeutic target for intestinal metabolic diseases.
Collapse
Affiliation(s)
- Yu Yang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jiakun Miao
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Juan Du
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shuang Xu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Kaiyi Zhang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Tianwen Wu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Cong Tao
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yanfang Wang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Meiying Fang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Shulin Yang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
3
|
D'Antongiovanni V, Fornai M, Colucci R, Nericcio A, Benvenuti L, Di Salvo C, Segnani C, Pierucci C, Ippolito C, Nemeth ZH, Haskó G, Bernardini N, Antonioli L, Pellegrini C. Enteric glial NLRP3 inflammasome contributes to gut mucosal barrier alterations in a mouse model of diet-induced obesity. Acta Physiol (Oxf) 2024:e14232. [PMID: 39287080 DOI: 10.1111/apha.14232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 08/12/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
AIM In the present study, we investigated the involvement of NLRP3 inflammasome in the intestinal epithelial barrier (IEB) changes associated with obesity, and its role in the interplay between enteric glia and intestinal epithelial cells (IECs). METHODS Wild-type C57BL/6J and NLRP3-KO (-/-) mice were fed with high-fat diet (HFD) or standard diet for 8 weeks. Colonic IEB integrity and inflammasome activation were assessed. Immunolocalization of colonic mucosal GFAP- and NLRP3-positive cells along with in vitro coculture experiments with enteric glial cells (EGCs) and IECs allowed to investigate the potential link between altered IEB, enteric gliosis, and NLRP3 activation. RESULTS HFD mice showed increased body weight, altered IEB integrity, increased GFAP-positive glial cells, and NLRP3 inflammasome hyperactivation. HFD-NLRP3-/- mice showed a lower increase in body weight, an improvement in IEB integrity and an absence of enteric gliosis. Coculture experiments showed that palmitate and lipopolysaccharide contribute to IEB damage and promote enteric gliosis with consequent hyperactivation of enteric glial NLRP3/caspase-1/IL-1β signaling. Enteric glial-derived IL-1β release exacerbates the IEB alterations. Such an effect was abrogated upon incubation with anakinra (IL-1β receptor antagonist) and with conditioned medium derived from silenced-NLRP3 glial cells. CONCLUSION HFD intake elicits mucosal enteric gliotic processes characterized by a hyperactivation of NLRP3/caspase-1/IL-1β signaling pathway, that contributes to further exacerbate the disruption of intestinal mucosal barrier integrity. However, we cannot rule out the contribution of NLRP3 inflammasome activation from other cells, such as immune cells, in IEB alterations associated with obesity. Overall, our results suggest that enteric glial NLRP3 inflammasome might represent an interesting molecular target for the development of novel pharmacological approaches aimed at managing the enteric inflammation and intestinal mucosal dysfunctions associated with obesity.
Collapse
Affiliation(s)
- Vanessa D'Antongiovanni
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Anna Nericcio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Laura Benvenuti
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Clelia Di Salvo
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Cristina Segnani
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Clarissa Pierucci
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chiara Ippolito
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Zoltan H Nemeth
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Surgery, Morristown Medical Center, Morristown, New Jersey, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York, USA
| | - Nunzia Bernardini
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Carolina Pellegrini
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
4
|
Grancieri M, de São José VPB, Toledo RCL, Verediano TA, Sant'Ana C, Lúcio HG, Gonzalez de Mejia E, Martino HSD. Effect of digested chia seed protein on the gut microbiota and colon morphology of mice fed a high-saturated-fat diet. Food Funct 2024; 15:9284-9297. [PMID: 39162475 DOI: 10.1039/d4fo02199a] [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: 08/21/2024]
Abstract
The present study aimed to investigate the effect of digested total protein (DTP) from chia seed on the gut microbiota and morphology of mice fed with a high-fat diet. Forty-four male C57BL/6 mice were divided into 4 groups: AIN (standard diet), HF (high-fat diet), AIN + DTP (standard diet supplemented with 400 mg of digested chia seed protein), and HF + DTP (high-fat diet supplemented with 400 mg of digested chia seed protein) during 8 weeks. Colon morphology, tight junction's gene expression, and gut microbiota composition were evaluated. The consumption of digested chia seed protein (DTP) increased the crypts width, longitudinal and circular muscular layer. Furthermore, the AIN + DTP group enhanced the expression of tight junction proteins, including occludin and claudin, while the AIN + DTP and HF + DTP groups increase the zonula occludens expression. The α-diversity analysis showed a reduction in bacterial dominance in the HF + DTP group. All the experimental groups were grouped in different cluster, showing differences in the microbiota community in the β-diversity analyzes. The Firmicutes/Bacteroidetes ratio did not differ among the groups. The genera Olsenella and Dubosiella were increased in the AIN + DTP group, but the Oscillospiraceae_unclassified was increased in the HF + DTP group. The Alistipes was increased, while the Roseburia and Akkermansia were decreased in the AIN + DTP and HF + DTP groups. Then, the consumption of DTP from chia seed improved the gut microbiota composition and mucosal integrity, counteracting the adverse effects of high-fat diet.
Collapse
Affiliation(s)
- Mariana Grancieri
- Department of Pharmacy and Nutrition, Federal University of Espírito Santo, Alegre, ES, 29500-000, Brazil
| | | | - Renata Celi Lopes Toledo
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, 36571-000, Brazil.
| | | | - Cintia Sant'Ana
- Department of Pharmacy and Nutrition, Federal University of Espírito Santo, Alegre, ES, 29500-000, Brazil
| | - Haira Guedes Lúcio
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, 36571-000, Brazil.
| | - Elvira Gonzalez de Mejia
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, Illinois, 61801, USA
| | | |
Collapse
|
5
|
Lefebvre C, Tiffay A, Breemeersch CE, Dreux V, Bôle-Feysot C, Guérin C, Breton J, Maximin E, Monnoye M, Déchelotte P, Douard V, Goichon A, Coëffier M. Sex-dependent effects of a high fat diet on metabolic disorders, intestinal barrier function and gut microbiota in mouse. Sci Rep 2024; 14:19835. [PMID: 39191839 DOI: 10.1038/s41598-024-70931-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
Obesity is often associated with sex-dependent metabolic complications, in which altered intestinal barrier function and gut microbiota contribute. We aimed to characterize in mice the sex-dependent effects of a high fat diet on these parameters. Male and female C57BL/6 mice received a standard (SD) or high fat diet (HFD; 60% kcal from fat) during 14 weeks (W14). Body composition, glucose tolerance, insulin sensitivity, intestinal permeability, colonic expression of 44 genes encoding factors involved in inflammatory response and gut barrier function, cecal microbiota, plasma adipokines and white adipose tissue response have been assessed. Both male and female HFD mice exhibited an increase of body weight and fat mass gain and glucose intolerance compared to SD mice. However, only male HFD mice tended to develop insulin resistance associated to increased Tnfα and Ccl2 mRNA expression in perigonadal adipose tissue. By contrast, only female HFD mice showed significant intestinal hyperpermeability that was associated with more markedly altered colonic inflammatory response. Cecal microbiota richness was markedly reduced in both sexes (Observed species) with sex-dependent modifications at the phyla or family level, e.g. decreased relative abundance of Bacillota and Lachnospiraceae in females, increased of Bacteroidaceae in males. Interestingly, some of these microbiota alterations were correlated with peripheral metabolic and inflammatory markers. In conclusions, male and female mice exhibit different responses to a high fat diet with specific changes of gut microbiota, intestinal barrier function, colonic and white adipose tissue inflammation, metabolic markers and body weight gain. The underlying mechanisms should be deciphered in further investigations.
Collapse
Affiliation(s)
- Candice Lefebvre
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis", UFR Santé, 22 Boulevard Gambetta, 76000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Univ Rouen Normandie, 76000, Rouen, France
| | - Adam Tiffay
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis", UFR Santé, 22 Boulevard Gambetta, 76000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Univ Rouen Normandie, 76000, Rouen, France
| | - Charles-Edward Breemeersch
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis", UFR Santé, 22 Boulevard Gambetta, 76000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Univ Rouen Normandie, 76000, Rouen, France
| | - Virginie Dreux
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis", UFR Santé, 22 Boulevard Gambetta, 76000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Univ Rouen Normandie, 76000, Rouen, France
| | - Christine Bôle-Feysot
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis", UFR Santé, 22 Boulevard Gambetta, 76000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Univ Rouen Normandie, 76000, Rouen, France
| | - Charlène Guérin
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis", UFR Santé, 22 Boulevard Gambetta, 76000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Univ Rouen Normandie, 76000, Rouen, France
| | - Jonathan Breton
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis", UFR Santé, 22 Boulevard Gambetta, 76000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Univ Rouen Normandie, 76000, Rouen, France
| | - Elise Maximin
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Domaine de Vilvert, 78350, Jouy-en-Josas, France
| | - Magali Monnoye
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Domaine de Vilvert, 78350, Jouy-en-Josas, France
| | - Pierre Déchelotte
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis", UFR Santé, 22 Boulevard Gambetta, 76000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Univ Rouen Normandie, 76000, Rouen, France
- Department of Nutrition, CHU Rouen, 76000, Rouen, France
| | - Véronique Douard
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Domaine de Vilvert, 78350, Jouy-en-Josas, France
| | - Alexis Goichon
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis", UFR Santé, 22 Boulevard Gambetta, 76000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Univ Rouen Normandie, 76000, Rouen, France
| | - Moïse Coëffier
- Univ Rouen Normandie, INSERM, Normandie Univ, ADEN UMR1073 "Nutrition, Inflammation and Microbiota-Gut-Brain Axis", UFR Santé, 22 Boulevard Gambetta, 76000, Rouen, France.
- Institute for Research and Innovation in Biomedicine (IRIB), Univ Rouen Normandie, 76000, Rouen, France.
- Department of Nutrition, CHU Rouen, 76000, Rouen, France.
| |
Collapse
|
6
|
Sukmak P, Kulworasreth P, Treveeravoot S, Arinno A, Anuwongworavet S, Wachiradejkul W, Kulworasreth P, Teansuk N, Thongnak L, Amonlerdpison D, Inchai J, Jakrachai C, Akrimajirachoote N, Aonbangkhen C, Muanprasat C, Poolsri W, Vaddhanaphuti CS, Pongkorpsakol P. Solanum melongena L. Extract Promotes Intestinal Tight Junction Re-Assembly via SIRT-1-Dependent Mechanisms. Mol Nutr Food Res 2024; 68:e2400230. [PMID: 39086054 DOI: 10.1002/mnfr.202400230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/30/2024] [Indexed: 08/02/2024]
Abstract
Tight junction disruption can lead to pathogenesis of various diseases without therapeutic strategy to recover intestinal barrier integrity. The main objective of this study is to demonstrate the effect of Solanum melongena L. extract (SMLE) on intestinal tight junction recovery and its underlying mechanism. Intestinal barrier function is attenuated by Ca2+ depletion. SMLE treatment increased TER value across T84 cell monolayers. Permeability assay reveals that Ca2+ depletion promotes 4-kDa FITC-dextran permeability, but not 70-kDa FITC-dextran. SMLE suppresses the rate of 4-kDa FITC-dextran permeability, indicating that SMLE inhibits paracellular leak pathway permeability. SMLE-mediated TER increase and leak pathway suppression are abolished by neither calcium/calmodulin-dependent protein kinase kinase β (CaMKKβ) inhibitor nor AMP-activated protein kinase (AMPK) inhibitor. Furthermore, mammalian target of rapamycin (mTOR) and extracellular signal-regulated kinase (ERK) inhibitors have no effects on SMLE-mediated TER increase and leak pathway suppression. Interestingly, SMLE is unable to enhance TER value and diminish leak pathway permeability in T84 cell monolayers pre-treated with sirtuin-1 (SIRT-1) inhibitor. Immunofluorescence staining reveals that SMLE enhances re-assembly of tight junction proteins, including occludin and ZO-1 to intercellular space but this effect is abolished by SIRT-1 inhibitor. These data suggest that SMLE promotes intestinal tight junction re-assembly via SIRT-1-dependent manner.
Collapse
Affiliation(s)
- Pichayapa Sukmak
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
- Laboratory of Epithelial Tight Junction Pathophysiology, Bangkok, Thailand
| | - Purisha Kulworasreth
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Supisara Treveeravoot
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
- Laboratory of Epithelial Tight Junction Pathophysiology, Bangkok, Thailand
| | - Apiwan Arinno
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
- Laboratory of Epithelial Tight Junction Pathophysiology, Bangkok, Thailand
- Center of Excellence in Natural Products Chemistry (CENP), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | | | - Wanapas Wachiradejkul
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Purit Kulworasreth
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Natnicha Teansuk
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
- Laboratory of Epithelial Tight Junction Pathophysiology, Bangkok, Thailand
| | - Laongdao Thongnak
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Doungporn Amonlerdpison
- Center of Excellence in Agricultural Innovation for Graduate Entrepreneur and Faculty of Fisheries Technology and Aquatic Resources, Maejo University, Chiang Mai, Thailand
| | - Jakkapong Inchai
- Innovative Research Unit of Epithelial Transport and Regulation (iETR), Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chaiwet Jakrachai
- Innovative Research Unit of Epithelial Transport and Regulation (iETR), Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Chanat Aonbangkhen
- Center of Excellence in Natural Products Chemistry (CENP), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | | | - Chutima S Vaddhanaphuti
- Innovative Research Unit of Epithelial Transport and Regulation (iETR), Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pawin Pongkorpsakol
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
- Laboratory of Epithelial Tight Junction Pathophysiology, Bangkok, Thailand
| |
Collapse
|
7
|
Bergheim I, Moreno-Navarrete JM. The relevance of intestinal barrier dysfunction, antimicrobial proteins and bacterial endotoxin in metabolic dysfunction-associated steatotic liver disease. Eur J Clin Invest 2024; 54:e14224. [PMID: 38634717 DOI: 10.1111/eci.14224] [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: 02/28/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is a leading cause of end-stage liver disease associated with increased mortality and cardiovascular disease. Obesity and diabetes are the most important risk factors of MASLD. It is well-established that obesity-associated insulin resistance leads to a situation of tissue lipotoxicity characterized by an accumulation of excess fat in non-fat tissues such as the liver, promoting the development of MASLD, and its progression into metabolic dysfunction-associated steatohepatitis. METHODS Here, we aimed to review the impact of disrupted intestinal permeability, antimicrobial proteins and bacterial endotoxin in the development and progression of MASLD. RESULTS AND CONCLUSION Recent studies demonstrated that obesity- and obesogenic diets-associated alterations of intestinal microbiota along with the disruption of intestinal barrier integrity, the alteration in antimicrobial proteins and, in consequence, an enhanced translocation of bacterial endotoxin into bloodstream might contribute to this pathological process through to impacting liver metabolism and inflammation.
Collapse
Affiliation(s)
- Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - José María Moreno-Navarrete
- Nutrition, Eumetabolism and Health Group, Institut d'Investigació Biomèdica de Girona (IDIBGI-CERCA), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Medicine, Universitat de Girona, Girona, Spain
| |
Collapse
|
8
|
Rini DM, Xu W, Suzuki T. Current Research on the Role of Isomaltooligosaccharides in Gastrointestinal Health and Metabolic Diseases. Prev Nutr Food Sci 2024; 29:93-105. [PMID: 38974594 PMCID: PMC11223922 DOI: 10.3746/pnf.2024.29.2.93] [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: 02/27/2024] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 07/09/2024] Open
Abstract
The intestinal epithelium plays an important role in maintaining the intestinal barrier and facilitating nutrient absorption. It also serves as a critical physical barrier against the infiltration of foreign substances from the intestinal lumen into the circulation. Intestinal barrier dysfunction has been implicated in the development of several diseases. Isomaltooligosaccharides (IMOs), which are a type of dietary fiber, possess multiple health benefits. However, there is limited information regarding their efficacy against gastrointestinal diseases. This review explores the therapeutic potential of IMOs in obesity, diabetes mellitus, inflammatory bowel disease (IBD), hyperlipidemia, and constipation. High-fat diet (HFD)-induced obesity models have shown that IMOs, administered alone or in combination with other compounds, exhibit potent antiobesity effects, making them promising agents in the treatment of obesity and its associated complications. Moreover, IMOs exhibit preventive effects against HFD-induced metabolic dysfunction by modulating gut microbiota and short-chain fatty acid levels, thereby ameliorating symptoms. Furthermore, IMOs can reduce IBD and alleviate hyperlipidemia, as indicated by the reduced histological colitis scores and improved lipid profiles observed in clinical trials and animal studies. This review highlights IMOs as a versatile intervention strategy that can improve gastrointestinal health by modulating gut microbiota, immune responses, and metabolic parameters, providing a multifaceted approach to address the complex nature of gastrointestinal disorders.
Collapse
Affiliation(s)
- Dina Mustika Rini
- Department of Food Technology, Faculty of Engineering, Universitas Pembangunan Nasional “Veteran” Jawa Timur, Surabaya 60294, Indonesia
| | - Wenxi Xu
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100193, China
| | - Takuya Suzuki
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan
| |
Collapse
|
9
|
Chmielarz M, Sobieszczańska B, Środa-Pomianek K. Metabolic Endotoxemia: From the Gut to Neurodegeneration. Int J Mol Sci 2024; 25:7006. [PMID: 39000116 PMCID: PMC11241432 DOI: 10.3390/ijms25137006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024] Open
Abstract
Metabolic endotoxemia is a severe health problem for residents in developed countries who follow a Western diet, disrupting intestinal microbiota and the whole organism's homeostasis. Although the effect of endotoxin on the human immune system is well known, its long-term impact on the human body, lasting many months or even years, is unknown. This is due to the difficulty of conducting in vitro and in vivo studies on the prolonged effect of endotoxin on the central nervous system. In this article, based on the available literature, we traced the path of endotoxin from the intestines to the blood through the intestinal epithelium and factors promoting the development of metabolic endotoxemia. The presence of endotoxin in the bloodstream and the inflammation it induces may contribute to lowering the blood-brain barrier, potentially allowing its penetration into the central nervous system; although, the theory is still controversial. Microglia, guarding the central nervous system, are the first line of defense and respond to endotoxin with activation, which may contribute to the development of neurodegenerative diseases. We traced the pro-inflammatory role of endotoxin in neurodegenerative diseases and its impact on the epigenetic regulation of microglial phenotypes.
Collapse
Affiliation(s)
- Mateusz Chmielarz
- Department of Microbiology, Wroclaw University of Medicine, Chalubinskiego 4 Street, 50-368 Wroclaw, Poland
| | - Beata Sobieszczańska
- Department of Microbiology, Wroclaw University of Medicine, Chalubinskiego 4 Street, 50-368 Wroclaw, Poland
| | - Kamila Środa-Pomianek
- Department of Biophysics and Neuroscience, Wroclaw University of Medicine, Chalubinskiego 3a, 50-368 Wroclaw, Poland
| |
Collapse
|
10
|
Tian W, Liu L, Wang R, Quan Y, Tang B, Yu D, Zhang L, Hua H, Zhao J. Gut microbiota in insulin resistance: a bibliometric analysis. J Diabetes Metab Disord 2024; 23:173-188. [PMID: 38932838 PMCID: PMC11196565 DOI: 10.1007/s40200-023-01342-x] [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: 08/20/2023] [Accepted: 11/06/2023] [Indexed: 06/28/2024]
Abstract
Background Insulin resistance (IR) is considered the pathogenic driver of diabetes, and can lead to obesity, hypertension, coronary artery disease, metabolic syndrome, and other metabolic disorders. Accumulating evidence indicates that the connection between gut microbiota and IR. This bibliometric analysis aimed to summarize the knowledge structure of gut microbiota in IR. Methods Articles and reviews related to gut microbiota in IR from 2013 to 2022 were retrieved from the Web of Science Core Collection (WoSCC), and the bibliometric analysis and visualization were performed by Microsoft Excel, Origin, R package (bibliometrix), Citespace, and VOSviewer. Results A total of 4 749 publications from WoSCC were retrieved, including 3 050 articles and 1 699 reviews. The majority of publications were from China and USA. The University Copenhagen and Shanghai Jiao Tong University were the most active institutions. The journal of Nutrients published the most papers, while Nature was the top 1 co-cited journal, and the major area of these publications was molecular, biology, and immunology. Nieuwdorp M published the highest number of papers, and Cani PD had the highest co-citations. Keyword analysis showed that the most frequently occurring keywords were "gut microbiota", "insulin-resistance", "obesity", and "inflammation". Trend topics and thematic maps showed that serum metabolome and natural products, such as resveratrol, flavonoids were the research hotspots in this field. Conclusion This bibliometric analysis summarised the hotspots, frontiers, pathogenesis, and treatment strategies, providing a clear and comprehensive profile of gut microbiota in IR. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-023-01342-x.
Collapse
Affiliation(s)
- Weiwei Tian
- Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Academy of Chinese Medical Sciences, Sichuan Institute for Translational Chinese Medicine, 610041 Chengdu, China
| | - Li Liu
- Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Academy of Chinese Medical Sciences, Sichuan Institute for Translational Chinese Medicine, 610041 Chengdu, China
| | - Ruirui Wang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Yunyun Quan
- Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Academy of Chinese Medical Sciences, Sichuan Institute for Translational Chinese Medicine, 610041 Chengdu, China
| | - Bihua Tang
- Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Academy of Chinese Medical Sciences, Sichuan Institute for Translational Chinese Medicine, 610041 Chengdu, China
| | - Dongmei Yu
- Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Academy of Chinese Medical Sciences, Sichuan Institute for Translational Chinese Medicine, 610041 Chengdu, China
| | - Lei Zhang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Hua Hua
- Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Academy of Chinese Medical Sciences, Sichuan Institute for Translational Chinese Medicine, 610041 Chengdu, China
| | - Junning Zhao
- Key Lab.: Biological Evaluation of TCM Quality of the State Administration of Traditional Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Academy of Chinese Medical Sciences, Sichuan Institute for Translational Chinese Medicine, 610041 Chengdu, China
| |
Collapse
|
11
|
Chae YR, Lee YR, Kim YS, Park HY. Diet-Induced Gut Dysbiosis and Leaky Gut Syndrome. J Microbiol Biotechnol 2024; 34:747-756. [PMID: 38321650 DOI: 10.4014/jmb.2312.12031] [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: 12/26/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/08/2024]
Abstract
Chronic gut inflammation promotes the development of metabolic diseases such as obesity. There is growing evidence which suggests that dysbiosis in gut microbiota and metabolites disrupt the integrity of the intestinal barrier and significantly impact the level of inflammation in various tissues, including the liver and adipose tissues. Moreover, dietary sources are connected to the development of leaky gut syndrome through their interaction with the gut microbiota. This review examines the effects of these factors on intestinal microorganisms and the communication pathways between the gut-liver and gut-brain axis. The consumption of diets rich in fats and carbohydrates has been found to weaken the adherence of tight junction proteins in the gastrointestinal tract. Consequently, this allows endotoxins, such as lipopolysaccharides produced by detrimental bacteria, to permeate through portal veins, leading to metabolic endotoxemia and alterations in the gut microbiome composition with reduced production of metabolites, such as short-chain fatty acids. However, the precise correlation between gut microbiota and alternative sweeteners remains uncertain, necessitating further investigation. This study highlights the significance of exploring the impact of diet on gut microbiota and the underlying mechanisms in the gut-liver and gut-brain axis. Nevertheless, limited research on the gut-liver axis poses challenges in comprehending the intricate connections between diet and the gut-brain axis. This underscores the need for comprehensive studies to elucidate the intricate gut-brain mechanisms underlying intestinal health and microbiota.
Collapse
Affiliation(s)
- Yu-Rim Chae
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do 55365, Republic of Korea
- Department of Food Science and Technology, Jeonbuk National University, Jeollabuk-do 54896, Republic of Korea
| | - Yu Ra Lee
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do 55365, Republic of Korea
| | - Young-Soo Kim
- Department of Food Science and Technology, Jeonbuk National University, Jeollabuk-do 54896, Republic of Korea
| | - Ho-Young Park
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do 55365, Republic of Korea
- Department of Food Biotechnology, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
12
|
Cai Y, Deng W, Yang Q, Pan G, Liang Z, Yang X, Li S, Xiao X. High-fat diet-induced obesity causes intestinal Th17/Treg imbalance that impairs the intestinal barrier and aggravates anxiety-like behavior in mice. Int Immunopharmacol 2024; 130:111783. [PMID: 38514921 DOI: 10.1016/j.intimp.2024.111783] [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: 11/08/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
The prevalence of autism spectrum disorders (ASD) has been steadily increasing, and growing evidence suggests a link between high-fat diet (HFD), obesity, and ASD; however, the mechanism underlying this association remains elusive. Herein, BTBR T + tf/J (BTBR) inbred mice (a mouse ASD model) and C57Bl/6J (C57) mice were fed an HFD and normal diet (ND) for 8 weeks (groups: C57 + ND, C57 + HFD, BTBR + ND, and BTBR + HFD). Subsequently, mice underwent behavioral assessments, followed by intestinal tissues harvesting to detect expression of intestinal barrier proteins and inflammatory factors and immune cell numbers, and a correlation analysis. HFD-fed BTBR mice developed obesity, elevated blood sugar, significantly aggravated anxiety-like behaviors, impaired intestinal barrier function, intestinal inflammation with elevated CD4+IL17+ T (Th17) cells and reduced CD4+Foxp3+ T (Treg) cells, exhibiting reduced expression of proteins related to AMPK regulatory pathway (AMPK, p-AMPK, SIRT1). Correlation analysis revealed that the degree of behavioral anxiety, the degree of intestinal barrier damage, the severity of intestinal inflammation, and the degree of immune cell imbalance positively correlated with each other. Accordingly, HFD-induced obesity may cause intestinal Th17/Treg imbalance via the AMPK-SIRT1 pathway, leading to an inflammatory environment in the intestine, impairing intestinal barrier function, and ultimately aggravating anxiety-like behaviors in mice.
Collapse
Affiliation(s)
- Yao Cai
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Wenlin Deng
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Qiuping Yang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Guixian Pan
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Zao Liang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Ximei Yang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China
| | - Sitao Li
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China.
| | - Xin Xiao
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510530, China.
| |
Collapse
|
13
|
Miranda CS, Santana-Oliveira DA, Vasques-Monteiro IL, Dantas-Miranda NS, Glauser JSDO, Silva-Veiga FM, Souza-Mello V. Time-dependent impact of a high-fat diet on the intestinal barrier of male mice. World J Methodol 2024; 14:89723. [PMID: 38577199 PMCID: PMC10989404 DOI: 10.5662/wjm.v14.i1.89723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/26/2023] [Accepted: 02/18/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Excessive saturated fat intake compromises the integrity of the intestinal mucosa, leading to low-grade inflammation, impaired mucosal integrity, and increased intestinal permeability, resulting in the migration of lipopolysaccharide (LPS) to other tissues. AIM To evaluate the chronic effects (at 10 and 16 wk) of a high-fat diet (HFD) (with 50% energy as fat) on the phylogenetic gut microbiota distribution and intestinal barrier structure and protection in C57BL/6 mice. METHODS Forty adult male mice were divided into four nutritional groups, where the letters refer to the type of diet (control and HFD or HF) and the numbers refer to the period (in weeks) of diet administration: Control diet for 10 wk, HFD for 10 wk, control diet for 16 wk, and HFD for 16 wk. After sacrifice, biochemical, molecular, and stereological analyses were performed. RESULTS The HF groups were overweight, had gut dysbiosis, had a progressive decrease in occludin immunostaining, and had increased LPS concentrations. Dietary progression reduced the number of goblet cells per large intestine area and Mucin2 expression in the HF16 group, consistent with a completely disarranged intestinal ultrastructure after 16 wk of HFD intake. CONCLUSION Chronic HFD intake causes overweight, gut dysbiosis, and morphological and functional alterations of the intestinal barrier after 10 or 16 wk. Time-dependent reductions in goblet cell numerical density and mucus production have emerged as targets for countering obesity-driven intestinal damage.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vanessa Souza-Mello
- Department of Anatomy, Rio de Janeiro State University, Rio de Janeiro 20551030, Brazil
| |
Collapse
|
14
|
Liu T, Zhao M, Zhang Y, Xu R, Fu Z, Jin T, Song J, Huang Y, Wang M, Zhao C. Polysaccharides from Phellinus linteus attenuate type 2 diabetes mellitus in rats via modulation of gut microbiota and bile acid metabolism. Int J Biol Macromol 2024; 262:130062. [PMID: 38340923 DOI: 10.1016/j.ijbiomac.2024.130062] [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: 07/06/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is the most prevalent metabolic disorder. Polysaccharides from Phellinus linteus (PLP) have been found to have anti-diabetes effects, but the mechanism has not been elucidated. The purpose of this study was to investigate the mechanism of PLP on T2DM through the gut microbiota and bile acids metabolism. The T2DM rat model was induced by a high-fat high-carbohydrate (HFHC) diet and streptozocin (30 mg/kg). We found that PLP ameliorated diabetes symptoms. Besides, PLP intervention increased the abundance of g_Bacteroides, g_Parabacteroides, and g_Alistioes, which are associated with the biosynthesis of short-chain fatty acids (SCFAs) and bile acids (BAs) metabolism. Meanwhile, untargeted and targeted metabolomics indicated that PLP could regulate the composition of BAs and increase the levels of SCFAs. Real-time quantitative PCR (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA) were performed to analyze the expression levels of BAs metabolism enzymes in the liver. Finally, the results of correlation analysis and Glucagon-like peptide-1 (GLP-1) showed that PLP stimulated the release of GLP-1 by regulating SCFAs and BAs. In conclusion, this study demonstrated that PLP can regulate gut microbiota and BAs metabolism to promote GLP-1 secretion, thereby increasing insulin release, decreasing blood glucose and attenuating T2DM.
Collapse
Affiliation(s)
- Tingting Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Min Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Yumeng Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Ruixiang Xu
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Zixuan Fu
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Tong Jin
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Jiaxi Song
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China
| | - Yihe Huang
- School of Public Health, Shenyang Medical College, Huanghe North Street 146, Shenyang, Liaoning Province, China
| | - Miao Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China.
| | - Chunjie Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning Province, China.
| |
Collapse
|
15
|
Frühbeck G, Gómez-Ambrosi J, Ramírez B, Becerril S, Rodríguez A, Mentxaka A, Valentí V, Moncada R, Reina G, Baixauli J, Casado M, Silva C, Escalada J, Catalán V. Decreased expression of the NLRP6 inflammasome is associated with increased intestinal permeability and inflammation in obesity with type 2 diabetes. Cell Mol Life Sci 2024; 81:77. [PMID: 38315242 PMCID: PMC10844155 DOI: 10.1007/s00018-024-05124-3] [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: 08/22/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND Obesity-associated dysfunctional intestinal permeability contributes to systemic chronic inflammation leading to the development of metabolic diseases. The inflammasomes constitute essential components in the regulation of intestinal homeostasis. We aimed to determine the impact of the inflammasomes in the regulation of gut barrier dysfunction and metabolic inflammation in the context of obesity and type 2 diabetes (T2D). METHODS Blood samples obtained from 80 volunteers (n = 20 normal weight, n = 21 OB without T2D, n = 39 OB with T2D) and a subgroup of jejunum samples were used in a case-control study. Circulating levels of intestinal damage markers and expression levels of inflammasomes as well as their main effectors (IL-1β and IL-18) and key inflammation-related genes were analyzed. The impact of inflammation-related factors, different metabolites and Akkermansia muciniphila in the regulation of inflammasomes and intestinal integrity genes was evaluated. The effect of blocking NLRP6 by using siRNA in inflammation was also studied. RESULTS Increased circulating levels (P < 0.01) of the intestinal damage markers endotoxin, LBP, and zonulin in patients with obesity decreased (P < 0.05) after weight loss. Patients with obesity and T2D exhibited decreased (P < 0.05) jejunum gene expression levels of NLRP6 and its main effector IL18 together with increased (P < 0.05) mRNA levels of inflammatory markers. We further showed that while NLRP6 was primarily localized in goblet cells, NLRP3 was localized in the intestinal epithelial cells. Additionally, decreased (P < 0.05) mRNA levels of Nlrp1, Nlrp3 and Nlrp6 in the small intestinal tract obtained from rats with diet-induced obesity were found. NLRP6 expression was regulated by taurine, parthenolide and A. muciniphila in the human enterocyte cell line CCL-241. Finally, a significant decrease (P < 0.01) in the expression and release of MUC2 after the knockdown of NLRP6 was observed. CONCLUSIONS The increased levels of intestinal damage markers together with the downregulation of NLRP6 and IL18 in the jejunum in obesity-associated T2D suggest a defective inflammasome sensing, driving to an impaired epithelial intestinal barrier that may regulate the progression of multiple obesity-associated comorbidities.
Collapse
Affiliation(s)
- Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain.
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Amaia Mentxaka
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
| | - Víctor Valentí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gabriel Reina
- Department of Microbiology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Jorge Baixauli
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Marcos Casado
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
| | - Camilo Silva
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Escalada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
16
|
Guerbette T, Rioux V, Bostoën M, Ciesielski V, Coppens-Exandier H, Buraud M, Lan A, Boudry G. Saturated fatty acids differently affect mitochondrial function and the intestinal epithelial barrier depending on their chain length in the in vitro model of IPEC-J2 enterocytes. Front Cell Dev Biol 2024; 12:1266842. [PMID: 38362040 PMCID: PMC10867211 DOI: 10.3389/fcell.2024.1266842] [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: 07/25/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
Introduction: Maintenance of the intestinal barrier mainly relies on the mitochondrial function of intestinal epithelial cells that provide ATP through oxidative phosphorylation (OXPHOS). Dietary fatty acid overload might induce mitochondrial dysfunction of enterocytes and may increase intestinal permeability as indicated by previous in vitro studies with palmitic acid (C16:0). Yet the impact of other dietary saturated fatty acids remains poorly described. Methods: To address this question, the in vitro model of porcine enterocytes IPEC-J2 was treated for 3 days with 250 µM of lauric (C12:0), myristic (C14:0), palmitic (C16:0) or stearic (C18:0) acids. Results and discussion: Measurement of the transepithelial electrical resistance, reflecting tight junction integrity, revealed that only C16:0 and C18:0 increased epithelial permeability, without modifying the expression of genes encoding tight junction proteins. Bioenergetic measurements indicated that C16:0 and C18:0 were barely β-oxidized by IPEC-J2. However, they rather induced significant OXPHOS uncoupling and reduced ATP production compared to C12:0 and C14:0. These bioenergetic alterations were associated with elevated mitochondrial reactive oxygen species production and mitochondrial fission. Although C12:0 and C14:0 treatment induced significant lipid storage and enhanced fusion of the mitochondrial network, it only mildly decreased ATP production without altering epithelial barrier. These results point out that the longer chain fatty acids C16:0 and C18:0 increased intestinal permeability, contrary to C12:0 and C14:0. In addition, C16:0 and C18:0 induced an important energy deprivation, notably via increased proton leaks, mitochondrial remodeling, and elevated ROS production in enterocytes compared to C12:0 and C14:0.
Collapse
Affiliation(s)
- Thomas Guerbette
- Institut NuMeCan, INRAE, INSERM, University Rennes, Rennes, France
| | - Vincent Rioux
- Institut NuMeCan, INRAE, INSERM, University Rennes, Rennes, France
- Institut Agro Rennes-Angers, Rennes, France
| | - Mégane Bostoën
- Institut NuMeCan, INRAE, INSERM, University Rennes, Rennes, France
| | - Vincent Ciesielski
- Institut NuMeCan, INRAE, INSERM, University Rennes, Rennes, France
- Institut Agro Rennes-Angers, Rennes, France
| | | | - Marine Buraud
- Institut NuMeCan, INRAE, INSERM, University Rennes, Rennes, France
| | - Annaïg Lan
- Institut NuMeCan, INRAE, INSERM, University Rennes, Rennes, France
- UMR PNCA, AgroParisTech, INRAE, Université Paris-Saclay, Paris, France
| | - Gaëlle Boudry
- Institut NuMeCan, INRAE, INSERM, University Rennes, Rennes, France
| |
Collapse
|
17
|
Hamjane N, Mechita MB, Nourouti NG, Barakat A. Gut microbiota dysbiosis -associated obesity and its involvement in cardiovascular diseases and type 2 diabetes. A systematic review. Microvasc Res 2024; 151:104601. [PMID: 37690507 DOI: 10.1016/j.mvr.2023.104601] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/24/2023] [Accepted: 09/02/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Obesity is a complex, multifactorial disease caused by various factors. Recently, the role of the gut microbiota in the development of obesity and its complications has attracted increasing interest. PURPOSE This article focuses on the mechanisms by which gut microbiota dysbiosis induces insulin resistance, type 2 diabetes, and cardiovascular diseases linked to obesity, highlighting the mechanisms explaining the role of gut microbiota dysbiosis-associated inflammation in the onset of these pathologies. METHODS A systematic study was carried out to understand and summarize the published results on this topic. More than 150 articles were included in this search, including different types of studies, consulted by an online search in English using various electronic search databases and predefined keywords related to the objectives of our study. RESULTS We have summarized the data from the articles consulted in this search, and we have found a major gut microbiota alteration in obesity, characterized by a specific decrease in butyrate-producing bacteria and the production of metabolites and components that lead to metabolic impairments and affect the progression of various diseases associated with obesity through distinct signaling pathways, including insulin resistance, type 2 diabetes, and cardiovascular diseases (CVD). We have also focused on the major role of inflammation as a link between gut microbiota dysbiosis and obesity-associated metabolic complications by explaining the mechanisms involved. CONCLUSION Gut microbiota dysbiosis plays a crucial role in the development of various obesity-related metabolic abnormalities, among them type 2 diabetes and CVD, and represents a major challenge for chronic disease prevention and health. Indeed, the intestinal microbiota appears to be a promising target for the nutritional or therapeutic management of these diseases.
Collapse
Affiliation(s)
- Nadia Hamjane
- Research Team in Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology of Tangier, Abdelmalek Essaadi University, Morocco.
| | - Mohcine Bennani Mechita
- Research Team in Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology of Tangier, Abdelmalek Essaadi University, Morocco
| | - Naima Ghailani Nourouti
- Research Team in Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology of Tangier, Abdelmalek Essaadi University, Morocco
| | - Amina Barakat
- Research Team in Biomedical Genomics and Oncogenetics, Faculty of Sciences and Technology of Tangier, Abdelmalek Essaadi University, Morocco
| |
Collapse
|
18
|
Lee YR, Lee HB, Oh MJ, Kim Y, Park HY. Thyme Extract Alleviates High-Fat Diet-Induced Obesity and Gut Dysfunction. Nutrients 2023; 15:5007. [PMID: 38068865 PMCID: PMC10708554 DOI: 10.3390/nu15235007] [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: 11/02/2023] [Revised: 11/24/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Prolonged intake of a high-fat diet (HFD) disturbs the composition of gut microbiota, contributing to the development of metabolic diseases, notably obesity and increased intestinal permeability. Thyme (Thymus vulgaris L.), an aromatic plant, is known for its several therapeutic properties. In this study, we explored the potential of thyme extract (TLE) to mitigate HFD-induced metabolic derangements and improve the gut environment. Eight-week-old C57BL/6 mice were administered 50 or 100 mg/kg TLE for eight weeks. Administration of 100 mg/kg TLE resulted in decreased weight gain and body fat percentage, alongside the regulation of serum biomarkers linked to obesity induced by a HFD. Moreover, TLE enhanced intestinal barrier function by increasing the expression of tight junction proteins and ameliorated colon shortening. TLE also altered the levels of various metabolites. Especially, when compared with a HFD, it was confirmed that 2-hydroxypalmitic acid and 3-indoleacrylic acid returned to normal levels after TLE treatment. Additionally, we investigated the correlation between fecal metabolites and metabolic parameters; deoxycholic acid displayed a positive correlation with most parameters, except for colon length. In contrast, hypoxanthine was negatively correlated with most parameters. These results suggest a promising role for thyme in ameliorating obesity and related gut conditions associated with a HFD.
Collapse
Affiliation(s)
- Yu Ra Lee
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.R.L.); (H.-B.L.); (M.-J.O.); (Y.K.)
| | - Hye-Bin Lee
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.R.L.); (H.-B.L.); (M.-J.O.); (Y.K.)
| | - Mi-Jin Oh
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.R.L.); (H.-B.L.); (M.-J.O.); (Y.K.)
| | - Yoonsook Kim
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.R.L.); (H.-B.L.); (M.-J.O.); (Y.K.)
| | - Ho-Young Park
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.R.L.); (H.-B.L.); (M.-J.O.); (Y.K.)
- Department of Food Biotechnology, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
19
|
Kang Y, Ren P, Shen X, Kuang X, Yang X, Liu H, Yan H, Yang H, Kang X, Ding Z, Luo X, Ma J, Yang Y, Fan W. A Newly Synbiotic Combination Alleviates Obesity by Modulating the Gut Microbiota-Fat Axis and Inhibiting the Hepatic TLR4/NF-κB Signaling Pathway. Mol Nutr Food Res 2023; 67:e2300141. [PMID: 37594720 DOI: 10.1002/mnfr.202300141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/11/2023] [Indexed: 08/19/2023]
Abstract
SCOPE Obesity has been recognized as a worldwide public health crisis, this is accompanied by dysregulation of the intestinal microbiota and upregulation of liver steatosis and adipose inflammation. Synbiotic as a novel alternative therapy for obesity have recently gained much attention. METHODS This study innovatively research the anti-obesity properties of a newly synbiotic composed of Lactobacillus acidophilus, Bifidobacterium infantis and konjac glucomannan oligosaccharides. RESULTS The synbiotic treatment can reduce body weight, fat mass, blood sugar, liver steatosis and adipose inflammation in obesity mice fed by high-fat diet (HFD). Meanwhile, synbiotic treatment activated brown adipose tissue and improve energy, glucose and lipid metabolism. In addition, synbiotic treatment not solely enhanced the protection of intestinal barrier, but also ameliorated gut microbiota dysbiosis directly by enhancing beneficial microbes and reducing potentially harmful bacteria. Furthermore, the microbiome phenotype and functional prediction showed that synbiotic treatment can improve the gut microbiota functions involving inflammatory state, immune response, metabolism and pathopoiesia. CONCLUSION The synbiotic may be an effective candidate treatment strategy for the clinical prevention and treatment of obesity and other associated metabolic diseases such as hyperlipidemia, nonalcoholic fatty liver diseases by alleviating inflammatory response, regulating energy metabolism and maintaining the balance of intestinal microecology.
Collapse
Affiliation(s)
- Yongbo Kang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Peng Ren
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xiaorong Shen
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xiaoyu Kuang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xiaodan Yang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Haixia Liu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Huan Yan
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Hao Yang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xing Kang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Zeyuan Ding
- Laboratory of Morphology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xuguang Luo
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jieqiong Ma
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Ying Yang
- Department of Endocrinology, Affiliated Hospital of Yunnan University, Kunming, Yunnan, 650021, China
| | - Weiping Fan
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| |
Collapse
|
20
|
Losol P, Sokolowska M, Hwang YK, Ogulur I, Mitamura Y, Yazici D, Pat Y, Radzikowska U, Ardicli S, Yoon JE, Choi JP, Kim SH, van de Veen W, Akdis M, Chang YS, Akdis CA. Epithelial Barrier Theory: The Role of Exposome, Microbiome, and Barrier Function in Allergic Diseases. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2023; 15:705-724. [PMID: 37957791 PMCID: PMC10643858 DOI: 10.4168/aair.2023.15.6.705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/18/2023] [Accepted: 10/07/2023] [Indexed: 11/15/2023]
Abstract
Allergic diseases are a major public health problem with increasing prevalence. These immune-mediated diseases are characterized by defective epithelial barriers, which are explained by the epithelial barrier theory and continuously emerging evidence. Environmental exposures (exposome) including global warming, changes and loss of biodiversity, pollution, pathogens, allergens and mites, laundry and dishwasher detergents, surfactants, shampoos, body cleaners and household cleaners, microplastics, nanoparticles, toothpaste, enzymes and emulsifiers in processed foods, and dietary habits are responsible for the mucosal and skin barrier disruption. Exposure to barrier-damaging agents causes epithelial cell injury and barrier damage, colonization of opportunistic pathogens, loss of commensal bacteria, decreased microbiota diversity, bacterial translocation, allergic sensitization, and inflammation in the periepithelial area. Here, we review scientific evidence on the environmental components that impact epithelial barriers and microbiome composition and their influence on asthma and allergic diseases. We also discuss the historical overview of allergic diseases and the evolution of the hygiene hypothesis with theoretical evidence.
Collapse
Affiliation(s)
- Purevsuren Losol
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Medical Research Center, Seoul National University, Seoul, Korea
- Department of Molecular Biology and Genetics, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yu-Kyoung Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Jeong-Eun Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jun-Pyo Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sae-Hoon Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Medical Research Center, Seoul National University, Seoul, Korea
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yoon-Seok Chang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Medical Research Center, Seoul National University, Seoul, Korea.
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
21
|
Yazici D, Ogulur I, Pat Y, Babayev H, Barletta E, Ardicli S, Bel Imam M, Huang M, Koch J, Li M, Maurer D, Radzikowska U, Satitsuksanoa P, Schneider SR, Sun N, Traidl S, Wallimann A, Wawrocki S, Zhakparov D, Fehr D, Ziadlou R, Mitamura Y, Brüggen MC, van de Veen W, Sokolowska M, Baerenfaller K, Nadeau K, Akdis M, Akdis CA. The epithelial barrier: The gateway to allergic, autoimmune, and metabolic diseases and chronic neuropsychiatric conditions. Semin Immunol 2023; 70:101846. [PMID: 37801907 DOI: 10.1016/j.smim.2023.101846] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/08/2023]
Abstract
Since the 1960 s, our health has been compromised by exposure to over 350,000 newly introduced toxic substances, contributing to the current pandemic in allergic, autoimmune and metabolic diseases. The "Epithelial Barrier Theory" postulates that these diseases are exacerbated by persistent periepithelial inflammation (epithelitis) triggered by exposure to a wide range of epithelial barrier-damaging substances as well as genetic susceptibility. The epithelial barrier serves as the body's primary physical, chemical, and immunological barrier against external stimuli. A leaky epithelial barrier facilitates the translocation of the microbiome from the surface of the afflicted tissues to interepithelial and even deeper subepithelial locations. In turn, opportunistic bacterial colonization, microbiota dysbiosis, local inflammation and impaired tissue regeneration and remodelling follow. Migration of inflammatory cells to susceptible tissues contributes to damage and inflammation, initiating and aggravating many chronic inflammatory diseases. The objective of this review is to highlight and evaluate recent studies on epithelial physiology and its role in the pathogenesis of chronic diseases in light of the epithelial barrier theory.
Collapse
Affiliation(s)
- Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Elena Barletta
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Swiss Institute of Bioinformatics (SIB), Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manal Bel Imam
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mengting Huang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Jana Koch
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Swiss Institute of Bioinformatics (SIB), Davos, Switzerland
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Debbie Maurer
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | | | - Stephan R Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Na Sun
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China
| | - Stephan Traidl
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Alexandra Wallimann
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sebastian Wawrocki
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Damir Zhakparov
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Danielle Fehr
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Reihane Ziadlou
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Marie-Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Katja Baerenfaller
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Swiss Institute of Bioinformatics (SIB), Davos, Switzerland
| | - Kari Nadeau
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland.
| |
Collapse
|
22
|
D'Antongiovanni V, Segnani C, Ippolito C, Antonioli L, Colucci R, Fornai M, Bernardini N, Pellegrini C. Pathological Remodeling of the Gut Barrier as a Prodromal Event of High-Fat Diet-Induced Obesity. J Transl Med 2023; 103:100194. [PMID: 37290605 DOI: 10.1016/j.labinv.2023.100194] [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: 02/17/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/10/2023] Open
Abstract
Intestinal barrier alterations represent a primum movens in obesity and related intestinal dysfunctions. However, whether gut barrier remodeling represents prodromal events in obesity before weight gain, metabolic alterations, and systemic inflammation remains unclear. Herein, we examined morphologic changes in the gut barrier in a mouse model of high-fat diet (HFD) since the earliest phases of diet assumption. C57BL/6J mice were fed with standard diet (SD) or HFD for 1, 2, 4, or 8 weeks. Remodeling of intestinal epithelial barrier, inflammatory infiltrate, and collagen deposition in the colonic wall was assessed by histochemistry and immunofluorescence analysis. Obese mice displayed increased body and epididymal fat weight along with increased plasma resistin, IL-1β, and IL-6 levels after 8 weeks of HFD. Starting from 1 week of HFD, mice displayed (1) a decreased claudin-1 expression in lining epithelial cells, (2) an altered mucus in goblet cells, (3) an increase in proliferating epithelial cells in colonic crypts, (4) eosinophil infiltration along with an increase in vascular P-selectin, and (5) deposition of collagen fibers. HFD intake is associated with morphologic changes in the large bowel at mucosal and submucosal levels. In particular, the main changes include alterations in the mucous layer and intestinal epithelial barrier integrity and activation of mucosal defense-enhanced fibrotic deposition. These changes represent early events occurring before the development of obesity condition that could contribute to compromising the intestinal mucosal barrier and functions, opening the way for systemic dissemination.
Collapse
Affiliation(s)
- Vanessa D'Antongiovanni
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Cristina Segnani
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chiara Ippolito
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nunzia Bernardini
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy; Interdepartmental Research Centre "Nutraceuticals and Food for Health," University of Pisa, Pisa, Italy.
| | - Carolina Pellegrini
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
23
|
Chen Y, Tseng SH, Chen CY, Tsai YH. Application of Intestinal Barrier Molecules in the Diagnosis of Acute Cellular Rejection After Intestinal Transplantation. Transpl Int 2023; 36:11595. [PMID: 37745643 PMCID: PMC10514359 DOI: 10.3389/ti.2023.11595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023]
Abstract
Diagnosing acute rejection after intestinal transplantation currently heavily relies on histopathological analysis of graft biopsies. However, the invasive risks associated with ileoscopic examination and the inaccessibility for biopsy after ileostomy closure hinder real-time detection of rejection responses. Molecules comprising the intestinal barrier have been identified as physiological and molecular biomarkers for various bowel conditions and systemic diseases. To investigate the potential of barrier function-related molecules in diagnosing rejection after intestinal transplantation, plasma samples were collected longitudinally from transplant recipients. The samples were categorized into "indeterminate for rejection (IND)" and "acute rejection (AR)" groups based on clinical diagnoses at each time point. The longitudinal association between plasma levels of these barrier function-related molecules and acute rejection was analyzed using the generalized estimating equations (GEE) method. Logistic GEE models revealed that plasma levels of claudin-3, occludin, sIgA, and zonulin were independent variables correlated with the clinical diagnosis of acute rejection. The subsequent prediction model demonstrated moderate ability in discriminating between IND and AR samples, with a sensitivity of 76.0%, specificity of 89.2%, and accuracy of 84.6%. In conclusion, monitoring plasma levels of claudin-3, occludin, sIgA, and zonulin shows great potential in aiding the diagnosis of acute rejection after intestinal transplantation.
Collapse
Affiliation(s)
- Yun Chen
- Department of Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan
| | - Sheng-Hong Tseng
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Yen Chen
- Medicine and Institute of Emergency and Critical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Association for the Study of Small Intestinal Diseases, Taoyuan, Taiwan
| | - Ya-Hui Tsai
- Department of Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan
| |
Collapse
|
24
|
Costa KA, Oliveira MCD, Cordeiro LMDS, Val CH, Machado FS, Fernandes SOA, Cardoso VN, Teixeira MM, Silveira ALM, Ferreira AVM. Effect of high-refined carbohydrate diet on intestinal integrity. Nutrition 2023; 113:112084. [PMID: 37354649 DOI: 10.1016/j.nut.2023.112084] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 06/26/2023]
Abstract
OBJECTIVES One of the leading causes of obesity is the consumption of excess nutrients. Obesity is characterized by adipose tissue expansion, chronic low-grade inflammation, and metabolic alterations. Although consumption of a high-fat diet has been demonstrated to be a diet-induced obesity model associated with gut disorders, the same effect is not well explored in a mild-obesity model induced by high-refined carbohydrate (HC) diet intake. The intestinal tract barrier comprises mucus, epithelial cells, tight junctions, immune cells, and gut microbiota. This system is susceptible to dysfunction by excess dietary components that could increase intestinal permeability and bacterial translocation. The aim of this study was to evaluate whether an HC diet and the alterations resulting from its intake are linked to small intestine changes. METHODS Male BALB/c mice were fed a chow or an HC diet for 8 wk. RESULTS Although differences in body weight gain were not observed between the groups, mice fed the HC diet showed increased adiposity associated with metabolic alterations. The interferon-γ expression and myeloperoxidase levels were increased in the small intestine in mice fed an HC diet. However, the intestinal villi length, the expression of tight junctions (zonula occludens-1 and claudin-4) and tumor necrosis factor-α cytokine, and the percentage of intraepithelial lymphocytes did not differ in the jejunum or ileum between the groups. We did not observe differences in intestinal permeability and bacterial translocation. CONCLUSION Metabolic alterations caused by consumption of an HC diet lead to a mild obesity state that does not necessarily involve significant changes in intestinal integrity.
Collapse
Affiliation(s)
- Kátia Anunciação Costa
- Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marina Chaves de Oliveira
- Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Cynthia Honorato Val
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabiana Simão Machado
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Valbert Nascimento Cardoso
- Department of Clinical Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Letícia Malheiros Silveira
- Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adaliene Versiani Matos Ferreira
- Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
25
|
Huang PX, Yeh CL, Yang SC, Shirakawa H, Chang CL, Chen LH, Chiu YS, Chiu WC. Rice Bran Supplementation Ameliorates Gut Dysbiosis and Muscle Atrophy in Ovariectomized Mice Fed with a High-Fat Diet. Nutrients 2023; 15:3514. [PMID: 37630706 PMCID: PMC10458250 DOI: 10.3390/nu15163514] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Rice bran, a byproduct of rice milling, is rich in fiber and phytochemicals and confers several health benefits. However, its effects on gut microbiota and obesity-related muscle atrophy in postmenopausal status remain unclear. In this study, we investigated the effects of rice bran on gut microbiota, muscle synthesis, and breakdown pathways in estrogen-deficient ovariectomized (OVX) mice receiving a high-fat diet (HFD). ICR female mice were divided into five groups: sham, OVX mice receiving control diet (OC); OVX mice receiving HFD (OH); OVX mice receiving control diet and rice bran (OR); and OVX mice receiving HFD and rice bran (OHR). After twelve weeks, relative muscle mass and grip strength were high in rice bran diet groups. IL-6, TNF-α, MuRf-1, and atrogin-1 expression levels were lower, and Myog and GLUT4 were higher in the OHR group. Rice bran upregulated the expression of occludin and ZO-1 (gut tight junction proteins). The abundance of Akkermansiaceae in the cecum was relatively high in the OHR group. Our finding revealed that rice bran supplementation ameliorated gut barrier dysfunction and gut dysbiosis and also maintained muscle mass by downregulating the expression of MuRf-1 and atrogin-1 (muscle atrophy-related factors) in HFD-fed OVX mice.
Collapse
Affiliation(s)
- Pei-Xin Huang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei City 11031, Taiwan; (P.-X.H.); (C.-L.Y.); (S.-C.Y.); (L.-H.C.); (Y.-S.C.)
| | - Chiu-Li Yeh
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei City 11031, Taiwan; (P.-X.H.); (C.-L.Y.); (S.-C.Y.); (L.-H.C.); (Y.-S.C.)
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei City 11031, Taiwan
| | - Suh-Ching Yang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei City 11031, Taiwan; (P.-X.H.); (C.-L.Y.); (S.-C.Y.); (L.-H.C.); (Y.-S.C.)
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei City 11031, Taiwan
| | - Hitoshi Shirakawa
- International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai 980-8572, Japan;
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki Aza Aoba, Aoba-ku, Sendai 980-8572, Japan
| | - Chao-Lin Chang
- Food Industry Research and Development Institute, Hsinchu 300193, Taiwan;
| | - Li-Hsin Chen
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei City 11031, Taiwan; (P.-X.H.); (C.-L.Y.); (S.-C.Y.); (L.-H.C.); (Y.-S.C.)
| | - Yen-Shuo Chiu
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei City 11031, Taiwan; (P.-X.H.); (C.-L.Y.); (S.-C.Y.); (L.-H.C.); (Y.-S.C.)
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Wan-Chun Chiu
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei City 11031, Taiwan; (P.-X.H.); (C.-L.Y.); (S.-C.Y.); (L.-H.C.); (Y.-S.C.)
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei City 11031, Taiwan
- Department of Nutrition, Wan Fang Hospital, Taipei Medical University, Taipei City 11696, Taiwan
| |
Collapse
|
26
|
Lee HB, Kim HR, Kang MC, Jeong D, Choi HD, Hong JS, Park HY. Structural characteristics of resistant starch-enriched rice during digestion and its effects on gut barrier function in high-fat induced obese mice. Food Res Int 2023; 170:113011. [PMID: 37316078 DOI: 10.1016/j.foodres.2023.113011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/17/2023] [Accepted: 05/18/2023] [Indexed: 06/16/2023]
Abstract
Dietary supplementation with indigestible carbohydrates is known to improve the gut environment and prevent obesity and inflammatory diseases by modulating the gut microbiota. In previous work, we established a method for the production of resistant starch (RS)-enriched high-amylose rice (R-HAR) using citric acid. The present study aimed to evaluate changes in structural characteristics during digestion of R-HAR and its effects on the gut health. A three-step in vitro digestion and fermentation model was used, then, RS content, scanning electron microscopy, and branch chain length distribution were analyzed during in vitro digestion. During the digestion of R-HAR, the RS content increased, and the structure was predicted to have a greater impact on the gut microbiota and gut environment. To study the intestinal health effects of R-HAR, its anti-inflammatory and gut barrier integrity activities were assayed in HFD-induced mice. Intake of R-HAR suppressed colonic shortening and inflammatory responses induced by HFD. Furthermore, R-HAR exhibited gut barrier protective activity with an increase in tight junction protein levels. We determined that R-HAR may be a potentially beneficial intestinal environment improver, which may have various implications in the food industry as rice.
Collapse
Affiliation(s)
- Hye-Bin Lee
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do 55365, Republic of Korea
| | - Ha Ram Kim
- Research Division of Strategic Food Technology, Korea Food Research Institute, Jeollabuk-do 55365, Republic of Korea
| | - Min-Cheol Kang
- Research Division of Strategic Food Technology, Korea Food Research Institute, Jeollabuk-do 55365, Republic of Korea
| | - Duyun Jeong
- Department of Food and Food Service Industry, Kyungpook National University, Sangju 37224, Republic of Korea
| | - Hee-Don Choi
- Research Division of Strategic Food Technology, Korea Food Research Institute, Jeollabuk-do 55365, Republic of Korea
| | - Jung Sun Hong
- Research Division of Strategic Food Technology, Korea Food Research Institute, Jeollabuk-do 55365, Republic of Korea
| | - Ho-Young Park
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do 55365, Republic of Korea; Department of Food Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
27
|
Horowitz A, Chanez-Paredes SD, Haest X, Turner JR. Paracellular permeability and tight junction regulation in gut health and disease. Nat Rev Gastroenterol Hepatol 2023:10.1038/s41575-023-00766-3. [PMID: 37186118 PMCID: PMC10127193 DOI: 10.1038/s41575-023-00766-3] [Citation(s) in RCA: 143] [Impact Index Per Article: 143.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2023] [Indexed: 05/17/2023]
Abstract
Epithelial tight junctions define the paracellular permeability of the intestinal barrier. Molecules can cross the tight junctions via two distinct size-selective and charge-selective paracellular pathways: the pore pathway and the leak pathway. These can be distinguished by their selectivities and differential regulation by immune cells. However, permeability increases measured in most studies are secondary to epithelial damage, which allows non-selective flux via the unrestricted pathway. Restoration of increased unrestricted pathway permeability requires mucosal healing. By contrast, tight junction barrier loss can be reversed by targeted interventions. Specific approaches are needed to restore pore pathway or leak pathway permeability increases. Recent studies have used preclinical disease models to demonstrate the potential of pore pathway or leak pathway barrier restoration in disease. In this Review, we focus on the two paracellular flux pathways that are dependent on the tight junction. We discuss the latest evidence that highlights tight junction components, structures and regulatory mechanisms, their impact on gut health and disease, and opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Arie Horowitz
- UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, Normandie University, Rouen, France
| | - Sandra D Chanez-Paredes
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xenia Haest
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Yu Z, Hao Q, Liu SB, Zhang QS, Chen XY, Li SH, Ran C, Yang YL, Teame T, Zhang Z, Zhou ZG. The positive effects of postbiotic (SWF concentration®) supplemented diet on skin mucus, liver, gut health, the structure and function of gut microbiota of common carp (Cyprinus carpio) fed with high-fat diet. FISH & SHELLFISH IMMUNOLOGY 2023; 135:108681. [PMID: 36921883 DOI: 10.1016/j.fsi.2023.108681] [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: 01/25/2023] [Revised: 02/25/2023] [Accepted: 03/11/2023] [Indexed: 06/18/2023]
Abstract
Postbiotics are an emerging research interest in recent years, which shows that metabolites, lysate extracts, cell wall components and even culture supernatants of probiotics can also exhibit significant prebiotic effects. In this study postbiotic stress worry free concentration® (SWFC) were prepared from the composition of culture supernatant of Cetobacterium somerae and Lactococcus lactis. The positive effects of SWFC supplemented diets on the growth performance, skin mucus, liver and gut health, and intestinal microbiota profile of Cyprinus carpio fed with high fat diets were investigated. 180 C. carpio with an average body weight of (3.01 ± 0.01) g were selected and randomly divided into three groups. They were fed with one of the three experimental diets supplemented with SWFC of 0 (control), 0.2 and 0.3 g/kg for 98 days, afterwards indexes were detected. The results revealed that, addition of SWFC had no significant effect on growth performance of C. carpio, while it can improve the health of the fish remarkably. In addition, SWFC improved mucosal C3, T-AOC, SOD activities, and decreased lipid peroxidation product MDA level, which were notably better than those in the control group (P < 0.05). In terms of the liver health systems, C. carpio fed on the diet supplemented with 0.2 g/kg of SWFC, showed significant improvement of the liver injured by HFD and reduce the contents of serum ALT and AST, and liver TAG (P < 0.05; P < 0.01). The expression of inflammation-related and lipid synthesis genes revealed that SWFC0.2 group could noteworthy enhance antioxidant capacity, reduced the expression of pro-inflammatory factors (TNF-α, IL-1β) and lipid synthesis genes (ACC, FAS, PPAR-β, PPAR-γ), and up-regulated the expression of anti-inflammatory factors (TGF-β). Additionally, intestinal morphology arose inflammatory cell infiltration, while intestinal integrity was better in SWFC groups compared with the control. Furthermore, the contents of serum LPS and LBP were remarkably lower in the SWFC0.2 group compared with the control (P < 0.01). The mRNA expression of genes related to gut health indicated that SWFC supplementation noteworthy up-regulated the expression of antioxidant (Nrf2, CAT, GPX), immune (Hepcidin, IL-10) and tight junction protein-related (ZO-1, Occludin). Simultaneously, the results of GF-zebrafish showed that the relative expression of anti-inflammatory genes (IL-1β, TGF-β) and antioxidant related genes (Nrf2, HO-1) were significantly up-regulated in SWFC groups. Data on intestinal microbiota profile verified that, at the phylum level, the abundance of Fusobacteria was remarkably elevated in the SWFC groups (P < 0.05), whereas the abundance of Firmicutes was declined noteworthy in SWFC0.2 and SWFC0.3 compared to the control group (P < 0.05; P < 0.01) respectively. At the genus level, the abundance of Cetobacterium in the SWFC groups were notably higher than those in the control group (P < 0.05), while the Vibrio content in the SWFC groups was significantly decreased (P < 0.05). PCoA result indicated that the intestinal microflora of SWFC0.2 group was abundant and diverse. Our results elucidate that dietary supplementation of SWFC protects C. carpio from HFD induced inflammatory response and oxidative stress, ameliorate skin mucus, liver and gut health, and improve the gut microbiota balance. Therefore, SWFC could be considered as an improving-fish-health additive, when supplemented to aquatic animal feed. With regards to how SWFC regulates the immunity and inflammatory responses and which signal transductions are involved remains unclear and more scientific evidences are needed to address these issues.
Collapse
Affiliation(s)
- Zhe Yu
- Sino-Norway Fish Gut Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Qiang Hao
- Sino-Norway Fish Gut Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Shu-Bin Liu
- Sino-Norway Fish Gut Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Qing-Shuang Zhang
- Sino-Norway Fish Gut Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Xing-Yu Chen
- Sino-Norway Fish Gut Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Sheng-Hui Li
- Sino-Norway Fish Gut Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Ya-Lin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Tsegay Teame
- Sino-Norway Fish Gut Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China; Tigray Agricultural Research Institute, Mekelle, Tigray, Ethiopia
| | - Zhen Zhang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| | - Zhi-Gang Zhou
- Sino-Norway Fish Gut Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| |
Collapse
|
29
|
Fan S, Chen S, Lin L. Research progress of gut microbiota and obesity caused by high-fat diet. Front Cell Infect Microbiol 2023; 13:1139800. [PMID: 36992691 PMCID: PMC10040832 DOI: 10.3389/fcimb.2023.1139800] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/28/2023] [Indexed: 03/15/2023] Open
Abstract
Obesity, a chronic metabolic disorder caused by an energy imbalance, has been increasingly prevalent and poses a global health concern. The multifactorial etiology of obesity includes genetics factors, high-fat diet, gut microbiota, and other factors. Among these factors, the implication of gut microbiota in the pathogenesis of obesity has been prominently acknowledged. This study endeavors to investigate the potential contribution of gut microbiota to the development of high-fat diet induced obesity, as well as the current state of probiotic intervention therapy research, in order to provide novel insights for the prevention and management of obesity.
Collapse
Affiliation(s)
- Shuyi Fan
- Scientific Research Department, Brain Hospital of Hunan Province, Second People’s Hospital of Hunan Province, Changsha, Hunan, China
- Department of Clinical Medicine, Xiamen Medical College, Xiamen, Fujian, China
| | - Suyun Chen
- Department of Clinical Medicine, Xiamen Medical College, Xiamen, Fujian, China
| | - Lin Lin
- Scientific Research Department, Brain Hospital of Hunan Province, Second People’s Hospital of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
30
|
Chen L, Mao M, Liu D, Liu W, Wang Y, Xie L, Deng Y, Lin Y, Xu Y, Zhong X, Cao W. HC067047 as a potent TRPV4 inhibitor repairs endotoxemia colonic injury. Int Immunopharmacol 2023; 116:109648. [PMID: 36706595 DOI: 10.1016/j.intimp.2022.109648] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/13/2022] [Accepted: 12/24/2022] [Indexed: 01/26/2023]
Abstract
Colonic injury causes severe inflammation during systemic infections in patients with endotoxemia. The prevention of colonic injury could effectively reduce the progression of endotoxemia. We investigated the protective effects and detailed mechanisms of the TRPV4 inhibitor HC067047 in the treatment of colonic injury caused by endotoxemia. An LPS-induced endotoxemia colonic injury model was used to assess the in vivo effects of HC067047. Colon slices were detected by hematoxylin and eosin (HE) staining and immunofluorescence assays. Spectrophotometry was used to determine the levels of MDA, calcium, GSH, and GSSG. Alterations in oxidative stress/mitophagy/inflammatory pyroptosis-related markers were evaluated by Q-PCR and western blot assays. HC067047 reduced the body weight loss and spleen weight index of endotoxemic mice and partly recovered the normal morphology of the colonic mucous layer. As an inhibitor of the calcium permeant cation channel, HC067047 suppressed the phosphorylation of the CAMKIIɑ protein and levels of MDA and calcium, upregulated the ratio of GSH/GSSG, shortened the expression of oxidative stress-related proteins, and enhanced the expression of the anti-oxidative protein CAT in damaged colon tissues. Additionally, HC067047 maintained normal mitochondrial functions in endotoxemia colons by promoting mitochondrial fusion and biosynthesis and suppressing mitochondrial fission and the PINK/Parkin/mitophagy pathway. HC067047 potently blocked inflammatory pyroptosis and protected the colonic tight junction barrier. HC067047 restores endotoxemia colons against oxidative stress, mitophagy, inflammatory pyroptosis, and colonic barrier dysfunction. Hence, HC067047 therapy may be potentially useful in the treatment of colonic injury in endotoxemia.
Collapse
Affiliation(s)
- Ling Chen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Mingli Mao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Dandan Liu
- Department of Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Wenjia Liu
- Department of Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yajuan Wang
- Department of Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Lihua Xie
- Department of Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yingcheng Deng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yi Lin
- Cancer Research Institute, Key Laboratory of Cancer Cellular and Molecular Pathology of Hunan Provincial, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yang Xu
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xiaolin Zhong
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Wenyu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
31
|
Shon WJ, Jung MH, Kim Y, Kang GH, Choi EY, Shin DM. Sugar-sweetened beverages exacerbate high-fat diet-induced inflammatory bowel disease by altering the gut microbiome. J Nutr Biochem 2023; 113:109254. [PMID: 36572070 DOI: 10.1016/j.jnutbio.2022.109254] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/10/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022]
Abstract
High-fat diets (HFDs) and frequent consumption of sugar-sweetened beverages (SSBs) are potential contributors to increasing inflammatory bowel disease (IBD) incidences. While HFDs have been implicated in mild intestinal inflammation, the role of sucrose in SSBs remains unclear. Therefore, we studied the role of SSBs in IBD pathogenesis in a mouse model and humans. C57BL6/J mice were given ad libitum access to a sucrose solution or plain water for 10 weeks, with or without an HFD. Interestingly, sucrose solution consumption alone did not induce gut inflammation in mice; however, when combined with an HFD, it dramatically increased the inflammation score, submucosal edema, and CD45+ cell infiltration. 16S ribosomal RNA gene-sequencing revealed that sucrose solution and HFD co-consumption significantly increased the relative abundance of IBD-related pathogenic bacteria when compared with HFD consumption. RNA sequencing and flow cytometry showed that co-consumption promoted pro-inflammatory cytokine and chemokine synthesis, dendritic-cell expansion, and IFN-γ+TNF-α+CD4+ and CD8+ T-cell activation. Fecal microbiota transplantation from HFD- and sucrose water-fed mice into gut-sterilized mice increased the susceptibility to dextran sulfate sodium-induced colitis in the recipient mice. Consistent herewith, high consumption of SSBs and animal fat-rich diets markedly increased systemic inflammation-associated IBD marker expression in humans. In conclusion, SSBs exacerbate HFD-induced colitis by triggering a shift of the gut microbiome into a pathobiome. Our findings provide new insights for the development of strategies aimed at preventing IBD.
Collapse
Affiliation(s)
- Woo-Jeong Shon
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Seoul, Republic of Korea; Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea
| | - Min Ho Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Younghoon Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Gyeong Hoon Kang
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea; Institute of Environment and Human Interface, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Dong-Mi Shin
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Seoul, Republic of Korea; Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea; Institute on Aging, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
32
|
The Gut-Prostate Axis: A New Perspective of Prostate Cancer Biology through the Gut Microbiome. Cancers (Basel) 2023; 15:cancers15051375. [PMID: 36900168 PMCID: PMC10000196 DOI: 10.3390/cancers15051375] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/03/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Obesity and a high-fat diet are risk factors associated with prostate cancer, and lifestyle, especially diet, impacts the gut microbiome. The gut microbiome plays important roles in the development of several diseases, such as Alzheimer's disease, rheumatoid arthritis, and colon cancer. The analysis of feces from patients with prostate cancer by 16S rRNA sequencing has uncovered various associations between altered gut microbiomes and prostate cancer. Gut dysbiosis caused by the leakage of gut bacterial metabolites, such as short-chain fatty acids and lipopolysaccharide results in prostate cancer growth. Gut microbiota also play a role in the metabolism of androgen which could affect castration-resistant prostate cancer. Moreover, men with high-risk prostate cancer share a specific gut microbiome and treatments such as androgen-deprivation therapy alter the gut microbiome in a manner that favors prostate cancer growth. Thus, implementing interventions aiming to modify lifestyle or altering the gut microbiome with prebiotics or probiotics may curtail the development of prostate cancer. From this perspective, the "Gut-Prostate Axis" plays a fundamental bidirectional role in prostate cancer biology and should be considered when screening and treating prostate cancer patients.
Collapse
|
33
|
Wang Y, Wang B, Zeng Z, Liu R, Tang L, Meng X, Li W. Bacillus amyloliquefaciens SC06 attenuated high-fat diet induced anxiety-like behavior and social withdrawal of male mice by improving antioxidant capacity, intestinal barrier function and modulating intestinal dysbiosis. Behav Brain Res 2023; 438:114172. [PMID: 36280009 DOI: 10.1016/j.bbr.2022.114172] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022]
Abstract
Anxiety-like behavior and social withdrawal induced by obesity and oxidative stress are significant health concerns in contemporary society. Our previously study found that Bacillus amyloliquefaciens SC06 (SC06) decreased the body weight of high-fat diet (HFD)-fed male mice and protected porcine intestinal epithelial cells against oxidative stress. The present study further investigated the effect of SC06 on HFD-induced obesity, anxiety-like behavior and social withdrawal of male mice and explored its mechanism. Results showed that SC06 significantly decreased HFD-induced obesity as evidenced by the decreased body weight, weight of liver and epididymal fat. Meanwhile, SC06 attenuated the anxiety-like behavior of HFD-fed male mice as illustrated by the more exploration time in both the open arms of elevated plus maze and the central area of open field and the reversed their social withdrawal tested in the three-chamber social choice task. SC06 also reduced reactive oxygen species (ROS) concentration and normalized the mitochondrial morphology in the hippocampus. SC06 reduced the systemic inflammation and increased the expression of intestinal tight junctions (ZO-1 and Claudin1). Furthermore, SC06 also altered the microbial diversity and composition, and decreased Firmicutes to Bacteroidetes ratio of HFD-fed male mice. These findings suggest SC06 attenuate HFD-induced anxiety-like behavior and social withdrawal of male mice by attenuating hippocampal oxidation stress, systemic inflammation, dysbiosis and improving intestinal barrier function.
Collapse
Affiliation(s)
- Yang Wang
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, 310058 Hangzhou, China; College of Animal Science and Technology, Qingdao Agricultural University, 266109 Qingdao, China
| | - Baikui Wang
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, 310058 Hangzhou, China
| | - Zhonghua Zeng
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, 310058 Hangzhou, China
| | - Rongrong Liu
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, 310058 Hangzhou, China
| | - Li Tang
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, 310058 Hangzhou, China
| | - Xiaolu Meng
- Department of Psychology, School of Medical Humanitarians, Guizhou Medical University, 550025 Guiyang, China.
| | - Weifen Li
- Key Laboratory of Molecular Animal Nutrition and Feed Sciences, College of Animal Science, Zhejiang University, 310058 Hangzhou, China.
| |
Collapse
|
34
|
Sheikh N, Shakeel S, Akhtar T, Khawar MB. Inimical impact of high-fat diet on expression of heme oxygenase-1, trace metals content, and associated intestinal histopathology. Toxicol Res (Camb) 2023; 12:26-32. [PMID: 36866220 PMCID: PMC9972815 DOI: 10.1093/toxres/tfac078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/22/2022] [Accepted: 10/13/2022] [Indexed: 12/23/2022] Open
Abstract
A high-fat diet (HFD) is one of the most prominent causative factors for obesity and metabolic inflammation. The effects of HFD overconsumption on intestinal histology, expression of haem oxygenase-1 (HO-1), and transferrin receptor-2 (TFR2) remain elusive. The present study was conducted to analyze the effect of HFD on these parameters. To develop the HFD-induced obese model, rat colonies were divided into 3 groups; the control group was reared on normal rat chow, whereas groups I and II were given HFD for 16 weeks. Hematoxylin and eosin (H & E) staining revealed marked epithelial changes, inflammatory cell infiltrates, and destruction of mucosal architecture in both experimental groups as compared to the control group. Sudan Black B staining showed a high triglyceride deposition in the intestinal mucosa of animals fed on HFD. Atomic absorption spectroscopy revealed a decrease in tissue copper (Cu) and selenium (Se) concentration in both HFD experimental groups. Whereas the cobalt (Co) and manganese (Mn) levels were comparable to controls. The mRNA expression levels of HO-1 and TFR2 were found to be significantly upregulated in HFD groups compared to the control group. Hence, HFD consumption leads to histopathological changes and altered gene expression in the rodent intestine. So, one should remove HFD from daily meals to avoid related metabolic complications.
Collapse
Affiliation(s)
- Nadeem Sheikh
- Cell and Molecular Biology Lab, Institute of Zoology, University of the Punjab, Q-A Campus, Lahore 54590, Pakistan
| | - Shakira Shakeel
- Cell and Molecular Biology Lab, Institute of Zoology, University of the Punjab, Q-A Campus, Lahore 54590, Pakistan
| | - Tasleem Akhtar
- Department of Pharmacology, University of Health Sciences Lahore, 54600, Pakistan
| | - Muhammad Babar Khawar
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal 51600, Pakistan
| |
Collapse
|
35
|
Kawaguchi S, Sakuraba H, Kikuchi H, Matsuki K, Hayashi Y, Ding J, Tanaka Y, Seya K, Matsumiya T, Hiraga H, Fukuda S, Sasaki K, Imaizumi T. Polygonum tinctorium leaf extract ameliorates high-fat diet-induced intestinal epithelial damage in mice. Exp Ther Med 2023; 25:112. [PMID: 36793327 PMCID: PMC9922942 DOI: 10.3892/etm.2023.11811] [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: 10/12/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
Dietary fat strongly influences the intestinal mucosal barrier, which protects against invading pathogenic bacteria. A high-fat diet (HFD) compromises the integrity of epithelial tight junctions (TJs) and reduces mucin production, leading to intestinal barrier disruption and metabolic endotoxemia. It has been shown that the active constituents of indigo plants can protect against intestinal inflammation; however, their protective role in HFD-induced intestinal epithelial damage remains unknown. The present study aimed to investigate the effects of Polygonum tinctorium leaf extract (indigo Ex) on HFD-induced intestinal damage in mice. Male C57BL6/J mice were fed a HFD and injected intraperitoneally with either indigo Ex or phosphate-buffered saline (PBS) for 4 weeks. The expression levels of TJ proteins, zonula occludens-1 and Claudin-1, were analyzed by immunofluorescence staining and western blotting. The colon mRNA expression levels of tumor necrosis factor-α, interleukin (IL)-12p40, IL-10 and IL-22 were measured by reverse transcription-quantitative PCR. The results revealed that indigo Ex administration attenuated the HFD-induced shortening of the colon. Colon crypt length was shown to be significantly greater in the indigo Ex-treated group mice compared with that in the PBS-treated group mice. Moreover, indigo Ex administration increased the number of goblet cells, and ameliorated the redistribution of TJ proteins. Notably, indigo Ex significantly increased the colon mRNA expression levels of IL-10. Indigo Ex displayed little effect on the gut microbial composition of HFD-fed mice. Taken together, these results suggested that indigo Ex may protect against HFD-induced epithelial damage. The leaves of indigo plants contain promising natural therapeutic compounds that could be used to treat obesity-associated intestinal damage and metabolic inflammation.
Collapse
Affiliation(s)
- Shogo Kawaguchi
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan,Correspondence to: Dr Shogo Kawaguchi, Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Hirotake Sakuraba
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Hidezumi Kikuchi
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Kota Matsuki
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Yudai Hayashi
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Jiangli Ding
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Yusuke Tanaka
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan,Department of Respiratory Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Kazuhiko Seya
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Tomoh Matsumiya
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Science, Hirosaki, Aomori 036-8564, Japan
| | - Hiroto Hiraga
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Shinsaku Fukuda
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Kenroh Sasaki
- Division of Pharmacognosy, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| | - Tadaatsu Imaizumi
- Department of Vascular Biology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| |
Collapse
|
36
|
Nie L, Yan Q, Zhang S, Cao Y, Zhou X. Duodenal Mucosa: A New Target for the Treatment of Type 2 Diabetes. Endocr Pract 2023; 29:53-59. [PMID: 36309189 DOI: 10.1016/j.eprac.2022.10.177] [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: 07/15/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE After a high-fat and high-sugar diet, the duodenal mucosa of rodents proliferate and trigger the signal of insulin resistance, which may be the cause of type 2 diabetes (T2D). In response to this phenomenon, researchers have designed the duodenal mucosal resurfacing (DMR) procedure, mainly through the hydrothermal ablation procedure, to restore the normal mucosal surface, thereby correcting this abnormal metabolic signal. This article aims to understand the changes in duodenum before and after the onset or treatment of T2D, and the potential mechanisms of DMR procedure. METHODS A literature search of PubMed and Web of Science was conducted using appropriate keywords. RESULTS Both animal and clinical studies have shown that the villus thickness, intestinal cells, glucose transporters, enteric nerves, and gut microbiota and their metabolites in the duodenum undergo corresponding changes before and after the onset or treatment of T2D. These changes may be related to the pathogenesis of T2D. DMR procedure may produce beneficial glycemic and hepatic metabolic effects by regulating these changes. CONCLUSION The duodenum is an important metabolic signaling center, and limiting nutrient exposure to this critical region will have powerful metabolic benefits. The DMR procedure may regulate glycemic and hepatic parameters through various mechanisms, which needs to be further confirmed by a large number of animal and clinical studies.
Collapse
Affiliation(s)
- LiJuan Nie
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - QianHua Yan
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shu Zhang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - YuTian Cao
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - XiQiao Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
37
|
Yao B, Pan B, Tian T, Su X, Zhang S, Li H, Li W, Wang Y, Lv S, Zhang Z. Baihu renshen decoction ameliorates type 2 diabetes mellitus in rats through affecting gut microbiota enhancing gut permeability and inhibiting TLR4/NF-κB-mediated inflammatory response. Front Cell Infect Microbiol 2022; 12:1051962. [PMID: 36439213 PMCID: PMC9691847 DOI: 10.3389/fcimb.2022.1051962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/25/2022] [Indexed: 10/24/2023] Open
Abstract
Baihu Rensheng decoction (BHRS) can effectively improve insulin resistance (IR) and decrease blood glucose in diabetic patients. However, its specific mechanism of action remains unclear. In this study, a type 2 diabetes mellitus (T2DM) rat model was established using a high-fat diet combined with streptozotocin (STZ) injection and treated with BHRS. Firstly, the therapeutic and anti-inflammatory effects of BHRS on T2DM were evaluated. Secondly, the effects of BHRS on gut permeability were evaluated and western blot was used to detect the changes of TLR4/NF-κB pathway-related protein expressions in liver. Finally, 16S rRNA sequencing was used to detect alteration of gut microbiota diversity and abundance in rats after BHRS treatment. Our results showed that BHRS could alleviate the hyperglycemia, hyperlipidemia, IR, and pathological changes of liver, pancreas, and kidney in T2DM rats. BHRS could also decrease the levels of pro-inflammatory cytokines and inhibit the oxidative stress. Immunohistochemistry showed BHRS could increase the expression tight junction-related proteins (ZO-1 and occludin) in colon. Besides, the level of LPS in serum was decreased after BHRS treatment. Western blot results showed that the protein expression of TLR4, MyD88 and the phosphorylation IκB, and NF-κBp65 were lowered after BHRS treatment. 16S rRNA sequencing showed that BHRS treatment altered the diversity of gut microbiotra and decreases the Firmicutes/Bacteroidetes (F to B) ratio at the phylum level. At the genus level, BHRS could increase the relative abundances of Lactobacillus, Blautia, and Anaerostipes and decrease the relative abundances of Allobaculum, Candidatus Saccharimonas, and Ruminococcus. In conclusion, our study revealed the various ameliorative effects of BHRS on T2DM, including improving the liver and kidney functions and alleviating the hyperglycemia, hyperlipidemia, pathological changes, oxidative stress and inflammatory response. The mechanisms of BHRS on T2DM are likely linked to the repair of gut barrier and the inhibition of TLR4/NF-κB-mediated inflammatory response and the improvement in the dysbiosis of gut microbiota.
Collapse
Affiliation(s)
- Bin Yao
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Baochao Pan
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Tian Tian
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Xiuhai Su
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Shufang Zhang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Hanzhou Li
- Graduate School, Chengde Medical University, Chengde, China
| | - Wendong Li
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Yuansong Wang
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Shuquan Lv
- Department of Endocrinology, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, China
| | - Zhaiyi Zhang
- College of Integrated Traditional Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
38
|
Wang R, Santos JM, Dufour JM, Stephens ER, Miranda JM, Washburn RL, Hibler T, Kaur G, Lin D, Shen CL. Ginger Root Extract Improves GI Health in Diabetic Rats by Improving Intestinal Integrity and Mitochondrial Function. Nutrients 2022; 14:4384. [PMID: 36297069 PMCID: PMC9611027 DOI: 10.3390/nu14204384] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/30/2022] [Accepted: 10/11/2022] [Indexed: 12/06/2022] Open
Abstract
Background Emerging research suggests hyperglycemia can increase intestinal permeability. Ginger and its bioactive compounds have been reported to benefit diabetic animals due to their anti-inflammatory and antioxidant properties. In this study, we revealed the beneficial effect of gingerol-enriched ginger (GEG) on intestinal health (i.e., barrier function, mitochondrial function, and anti-inflammation) in diabetic rats. Methods Thirty-three male Sprague Dawley rats were assigned to three groups: low-fat diet (control group), high-fat-diet (HFD) + streptozotocin (single low dose 35 mg/kg body weight (BW) after 2 weeks of HFD feeding) (DM group), and HFD + streptozotocin + 0.75% GEG in diet (GEG group) for 42 days. Glucose tolerance tests (GTT) and insulin tolerance tests (ITT) were conducted at baseline and prior to sample collection. Total pancreatic insulin content was determined by ELISA. Total RNA of intestinal tissues was extracted for mRNA expression using qRT-PCR. Results Compared to the DM group, the GEG group had improved glucose tolerance and increased pancreatic insulin content. Compared to those without GEG (DM group), GEG supplementation (GEG group) increased the gene expression of tight junction (Claudin-3) and antioxidant capacity (SOD1), while it decreased the gene expression for mitochondrial fusion (MFN1), fission (FIS1), biogenesis (PGC-1α, TFAM), mitophagy (LC3B, P62, PINK1), and inflammation (NF-κB). Conclusions Ginger root extract improved glucose homeostasis in diabetic rats, in part, via improving intestinal integrity and mitochondrial dysfunction of GI health.
Collapse
Affiliation(s)
- Rui Wang
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Julianna Maria Santos
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Jannette M. Dufour
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79401, USA
| | - Emily R. Stephens
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Jonathan M. Miranda
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Rachel L. Washburn
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Taylor Hibler
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Gurvinder Kaur
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79401, USA
| | - Dingbo Lin
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Chwan-Li Shen
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79401, USA
| |
Collapse
|
39
|
Unhealthy Diets Induce Distinct and Regional Effects on Intestinal Inflammatory Signalling Pathways and Long-Lasting Metabolic Dysfunction in Rats. Int J Mol Sci 2022; 23:ijms231810984. [PMID: 36142897 PMCID: PMC9503261 DOI: 10.3390/ijms231810984] [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: 07/29/2022] [Revised: 09/09/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
The intestinal epithelium is a principal site for environmental agents’ detection. Several inflammation- and stress-related signalling pathways have been identified as key players in these processes. However, it is still unclear how the chronic intake of inadequate nutrients triggers inflammatory signalling pathways in different intestinal regions. We aimed to evaluate the impact of unhealthy dietary patterns, starting at a younger age, and the association with metabolic dysfunction, intestinal inflammatory response, and obesity in adulthood. A rat model was used to evaluate the effects of the consumption of sugary beverages (HSD) and a Western diet (WD), composed of ultra-processed foods. Both diets showed a positive correlation with adiposity index, but a positive correlation was found between the HSD diet and the levels of blood glucose and triglycerides, whereas the WD diet correlated positively with triglyceride levels. Moreover, a distinct inflammatory response was associated with either the WD or HSD diets. The WD induced an increase in TLR2, TLR4, and nuclear factor-kappa B (NF-κB) intestinal gene expression, with higher levels in the colon and overexpression of the inducible nitric oxide synthase. In turn, the HSD diet induced activation of the TLR2-mediated NF-κB signalling pathway in the small intestine. Altogether, these findings support the concept that early intake of unhealthy foods and nutrients are a main exogenous signal for disturbances of intestinal immune mechanisms and in a region-specific manner, ultimately leading to obesity-related disorders in later life.
Collapse
|
40
|
Wu Y, Liu X, Li J, Ji W, Tian Q, Li S. Aerobic exercise improves intestinal mucosal barrier dysfunction through TLR4/MyD88/NF-κB signaling pathway in diabetic rats. Biochem Biophys Res Commun 2022; 634:75-82. [DOI: 10.1016/j.bbrc.2022.09.075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 09/12/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022]
|
41
|
Cuddihey H, Cavin JB, Keenan CM, Wallace LE, Vemuri K, Makriyannis A, MacNaughton WK, Sharkey KA. Role of CB 1 receptors in the acute regulation of small intestinal permeability: effects of high-fat diet. Am J Physiol Gastrointest Liver Physiol 2022; 323:G219-G238. [PMID: 35787179 PMCID: PMC9394780 DOI: 10.1152/ajpgi.00341.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 01/31/2023]
Abstract
The endocannabinoid system of the gastrointestinal tract is involved in the control of intestinal barrier function. Whether the cannabinoid 1 (CB1) receptor is expressed on the intestinal epithelium and acutely regulates barrier function has not been determined. Here, we tested the hypothesis that ligands of the CB1 receptor acutely modulate small intestinal permeability and that this is associated with altered distribution of tight junction proteins. We examined the acute effects of CB1 receptor ligands on small intestinal permeability both in chow-fed and 2-wk high-fat diet (HFD)-fed mice using Ussing chambers. We assessed the distribution of CB1 receptor and tight junction proteins using immunofluorescence and the expression of CB1 receptor using PCR. A low level of CB1 expression was found on the intestinal epithelium. CB1 receptor was highly expressed on enteric nerves in the lamina propria. Neither the CB1/CB2 agonist CP55,940 nor the CB1 neutral antagonist AM6545 altered the flux of 4kDa FITC dextran (FD4) across the jejunum or ileum of chow-fed mice. Remarkably, both CP55,940 and AM6545 reduced FD4 flux across the jejunum and ileum in HFD-fed mice that have elevated baseline intestinal permeability. These effects were absent in CB1 knockout mice. CP55,940 reduced the expression of claudin-2, whereas AM6545 had little effect on claudin-2 expression. Neither ligand altered the expression of ZO-1. Our data suggest that CB1 receptor on the intestinal epithelium regulates tight junction protein expression and restores barrier function when it is increased following exposure to a HFD for 2 wk.NEW & NOTEWORTHY The endocannabinoid system of the gastrointestinal tract regulates homeostasis by acting as brake on motility and secretion. Here we show that when exposed to a high fat diet, intestinal permeability is increased and activation of the CB1 receptor on the intestinal epithelium restores barrier function. This work further highlights the role of the endocannabinoid system in regulating intestinal homeostasis when it is perturbed.
Collapse
Affiliation(s)
- Hailey Cuddihey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jean-Baptiste Cavin
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Catherine M Keenan
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Laurie E Wallace
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kiran Vemuri
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts
| | - Alexandros Makriyannis
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts
| | - Wallace K MacNaughton
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keith A Sharkey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
42
|
Abot A, Fried S, Cani PD, Knauf C. Reactive Oxygen Species/Reactive Nitrogen Species as Messengers in the Gut: Impact on Physiology and Metabolic Disorders. Antioxid Redox Signal 2022; 37:394-415. [PMID: 34714099 DOI: 10.1089/ars.2021.0100] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significance: The role of reactive oxygen/nitrogen species as "friend" or "foe" messengers in the whole body is well characterized. Depending on the concentration in the tissue considered, these molecular actors exert beneficial or deleterious impacts leading to a pathological state, as observed in metabolic disorders such as type 2 diabetes and obesity. Recent Advances: Among the tissues impacted by oxidation and inflammation in this pathological state, the intestine is a site of dysfunction that can establish diabetic symptoms, such as alterations in the intestinal barrier, gut motility, microbiota composition, and gut/brain axis communication. In the intestine, reactive oxygen/nitrogen species (from the host and/or microbiota) are key factors that modulate the transition from physiological to pathological signaling. Critical Issues: Controlling the levels of intestinal reactive oxygen/nitrogen species is a complicated balance between positive and negative impacts that is in constant equilibrium. Here, we describe the synthesis and degradation of intestinal reactive oxygen/nitrogen species and their interactions with the host. The development of novel redox-based therapeutics that alter these processes could restore intestinal health in patients with metabolic disorders. Future Directions: Deciphering the mode of action of reactive oxygen/nitrogen species in the gut of obese/diabetic patients could result in a future therapeutic strategy that combines nutritional and pharmacological approaches. Consequently, preventive and curative treatments must take into account one of the first sites of oxidative and inflammatory dysfunctions in the body, that is, the intestine. Antioxid. Redox Signal. 37, 394-415.
Collapse
Affiliation(s)
- Anne Abot
- Université Paul Sabatier, Toulouse III, INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan, Toulouse, France.,International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France
| | - Steven Fried
- Université Paul Sabatier, Toulouse III, INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan, Toulouse, France.,International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France
| | - Patrice D Cani
- International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France.,UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, WELBIO, Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Brussels, Belgium
| | - Claude Knauf
- Université Paul Sabatier, Toulouse III, INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan, Toulouse, France.,International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France
| |
Collapse
|
43
|
Urbonaite G, Knyzeliene A, Bunn FS, Smalskys A, Neniskyte U. The impact of maternal high-fat diet on offspring neurodevelopment. Front Neurosci 2022; 16:909762. [PMID: 35937892 PMCID: PMC9354026 DOI: 10.3389/fnins.2022.909762] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/27/2022] [Indexed: 12/11/2022] Open
Abstract
A maternal high-fat diet affects offspring neurodevelopment with long-term consequences on their brain health and behavior. During the past three decades, obesity has rapidly increased in the whole human population worldwide, including women of reproductive age. It is known that maternal obesity caused by a high-fat diet may lead to neurodevelopmental disorders in their offspring, such as autism spectrum disorder, attention deficit hyperactivity disorder, anxiety, depression, and schizophrenia. A maternal high-fat diet can affect offspring neurodevelopment due to inflammatory activation of the maternal gut, adipose tissue, and placenta, mirrored by increased levels of pro-inflammatory cytokines in both maternal and fetal circulation. Furthermore, a maternal high fat diet causes gut microbial dysbiosis further contributing to increased inflammatory milieu during pregnancy and lactation, thus disturbing both prenatal and postnatal neurodevelopment of the offspring. In addition, global molecular and cellular changes in the offspring's brain may occur due to epigenetic modifications including the downregulation of brain-derived neurotrophic factor (BDNF) expression and the activation of the endocannabinoid system. These neurodevelopmental aberrations are reflected in behavioral deficits observed in animals, corresponding to behavioral phenotypes of certain neurodevelopmental disorders in humans. Here we reviewed recent findings from rodent models and from human studies to reveal potential mechanisms by which a maternal high-fat diet interferes with the neurodevelopment of the offspring.
Collapse
Affiliation(s)
- Gintare Urbonaite
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Agne Knyzeliene
- Centre for Cardiovascular Science, The Queen’s Medical Research Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Fanny Sophia Bunn
- Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| | - Adomas Smalskys
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Urte Neniskyte
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- VU LSC-EMBL Partnership for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
44
|
Phillippi DT, Daniel S, Nguyen KN, Penaredondo BA, Lund AK. Probiotics Function as Immunomodulators in the Intestine in C57Bl/6 Male Mice Exposed to Inhaled Diesel Exhaust Particles on a High-Fat Diet. Cells 2022; 11:cells11091445. [PMID: 35563751 PMCID: PMC9101602 DOI: 10.3390/cells11091445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/10/2022] [Accepted: 04/20/2022] [Indexed: 12/04/2022] Open
Abstract
Epidemiological studies reveal a correlation between air pollution exposure and gastrointestinal (GI) diseases, yet few studies have investigated the role of inhaled particulate matter on intestinal integrity in conjunction with a high-fat (HF) diet. Additionally, there is currently limited information on probiotics in mitigating air-pollutant responses in the intestines. Thus, we investigated the hypothesis that exposure to inhaled diesel exhaust particles (DEP) and a HF diet can alter intestinal integrity and inflammation, which can be attenuated with probiotics. 4-6-w-old male C57Bl/6 mice on a HF diet (45% kcal fat) were randomly assigned to be exposed via oropharyngeal aspiration to 35 µg of DEP suspended in 35 µL of 0.9% sterile saline or sterile saline (CON) only twice a week for 4 w. A subset of mice was treated with 0.3 g/day of Winclove Ecologic® barrier probiotics (PRO) in drinking water throughout the duration of the study. Our results show that DEP exposure ± probiotics resulted in increased goblet cells and mucin (MUC)-2 expression, as determined by AB/PAS staining. Immunofluorescent quantification and/or RT-qPCR showed that DEP exposure increases claudin-3, occludin, zona occludens (ZO)-1, matrix metalloproteinase (MMP)-9, and toll-like receptor (TLR)-4, and decreases tumor necrosis factor (TNF)-α and interleukin (IL)-10 expression compared to CON. DEP exposure + probiotics increases expression of claudin-3, occludin, ZO-1, TNF-α, and IL-10 and decreases MMP-9 and TLR-4 compared to CON + PRO in the small intestine. Collectively, these results show that DEP exposure alters intestinal integrity and inflammation in conjunction with a HF diet. Probiotics proved fundamental in understanding the role of the microbiome in protecting and altering inflammatory responses in the intestines following exposure to inhaled DEP.
Collapse
Affiliation(s)
| | | | | | | | - Amie K. Lund
- Correspondence: ; Tel.: +1-(940)-369-8946; Fax: +1-(940)-565-4297
| |
Collapse
|
45
|
Collares-Buzato CB, Carvalho CP. Is type 2 diabetes mellitus another intercellular junction-related disorder? Exp Biol Med (Maywood) 2022; 247:743-755. [PMID: 35466731 DOI: 10.1177/15353702221090464] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Type 2 diabetes mellitus (T2D) is nowadays a worldwide epidemic and has become a major challenge for health systems around the world. It is a multifactorial disorder, characterized by a chronic state of hyperglycemia caused by defects in the production as well as in the peripheral action of insulin. This minireview highlights the experimental and clinical evidence that supports the novel idea that intercellular junctions (IJs)-mediated cell-cell contacts play a role in the pathogenesis of T2D. It focuses on IJs repercussion for endocrine pancreas, intestinal barrier, and kidney dysfunctions that contribute to the onset and evolution of this metabolic disorder.
Collapse
Affiliation(s)
- Carla B Collares-Buzato
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, CEP 13083-970, Brazil
| | - Carolina Pf Carvalho
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, SP, CEP 11015-020, Brazil
| |
Collapse
|
46
|
Almeida PP, Valdetaro L, Thomasi BBDM, Stockler-Pinto MB, Tavares-Gomes AL. High-fat diets on the enteric nervous system: Possible interactions and mechanisms underlying dysmotility. Obes Rev 2022; 23:e13404. [PMID: 34873814 DOI: 10.1111/obr.13404] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/25/2021] [Accepted: 11/15/2021] [Indexed: 01/09/2023]
Abstract
Obesity is a chronic disease that affects various physiological systems. Among them, the gastrointestinal tract appears to be a main target of this disease. High-fat diet (HFD) animal models can help recapitulate the classic signs of obesity and present a series of gastrointestinal alterations, mainly dysmotility. Because intestinal motility is governed by the enteric nervous system (ENS), enteric neurons, and glial cells have been studied in HFD models. Given the importance of the ENS in general gut physiology, this review aims to discuss the relationship between HFD-induced neuroplasticity and gut dysmotility observed in experimental models. Furthermore, we highlight components of the gut environment that might influence enteric neuroplasticity, including gut microbiota, enteric glio-epithelial unit, serotonin release, immune cells, and disturbances such as inflammation and oxidative stress.
Collapse
Affiliation(s)
| | - Luisa Valdetaro
- Postgraduate Program in Neurosciences, Fluminense Federal University, Niterói, Brazil
| | | | - Milena Barcza Stockler-Pinto
- Postgraduate Program in Cardiovascular Sciences, Fluminense Federal University, Niterói, Brazil.,Postgraduate Program in Nutrition Sciences, Fluminense Federal University, Niterói, Brazil
| | | |
Collapse
|
47
|
Zhang FL, Hao Q, Zhang QS, Lv HY, Yang YL, Zhang Z, Zhou ZG. Influences of dietary Eucommia ulmoides leaf extract on the hepatic lipid metabolism, inflammation response, intestinal antioxidant capacity, intestinal microbiota, and disease resistance of the channel catfish (Ictalurus punctatus). FISH & SHELLFISH IMMUNOLOGY 2022; 123:75-84. [PMID: 35240294 DOI: 10.1016/j.fsi.2022.02.053] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/24/2022] [Accepted: 02/24/2022] [Indexed: 06/14/2023]
Abstract
The purpose of the study was to investigate the effects of Eucommia ulmoides leaf extract (ELE) on the common occurrence of liver steatosis, chronic inflammation, oxidative stress, disturbance of gut microbiota, and disease susceptibility in high-fat diet-fed channel catfish. Channel catfish fed three diets, including a high-fat diet (11% crude fat) and ELE-supplemented diets containing 1‰ or 2‰ ELE for 4 weeks. The results showed the contents of liver triacylglycerol of 1‰ and 2‰ ELE groups were reduced, and ELE treatments decreased the expression of lipogenesis related genes (srebp-1c, pparγ, and acc-1), and increased the expression of lipolysis related genes (pparα). In addition, the supplementation of ELE improved the inflammatory response of the liver and intestine. ELE could improve the destruction of intestinal morphology structure and increase the expression level of hif-1a and tight junction proteins (Occludin, Claudin2, Claudin15). 2‰ ELE significantly enhanced the antioxidant capacity of intestine by increasing the activity of SOD enzyme. Moreover, the supplement of ELE significantly increased the abundance of Cetobacterium and Romboutsia (p < 0.05). Compared with the control group, the expression of immune factor nf-κb had a significant decrease, and il-1β showed a tendency to decrease in the ELE supplement groups after pathogenic bacteria challenge. In conclusion, the ELE alleviated fatty liver disease and inflammation response, improved the oxidative capacity and physiological structure of intestine, and improved the structure of intestinal microbiota and disease resistance in HFD-fed channel catfish.
Collapse
Affiliation(s)
- Feng-Li Zhang
- Sino-Norway Fish Gastrointestinal Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Qiang Hao
- Sino-Norway Fish Gastrointestinal Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Qing-Shuang Zhang
- Sino-Norway Fish Gastrointestinal Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Hui-Yuan Lv
- Beijing CENTRE Technology, Beijing, 100081, China
| | - Ya-Lin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Zhen Zhang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| | - Zhi-Gang Zhou
- Sino-Norway Fish Gastrointestinal Microbiota Joint Lab, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| |
Collapse
|
48
|
Micronutrient Improvement of Epithelial Barrier Function in Various Disease States: A Case for Adjuvant Therapy. Int J Mol Sci 2022; 23:ijms23062995. [PMID: 35328419 PMCID: PMC8951934 DOI: 10.3390/ijms23062995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
The published literature makes a very strong case that a wide range of disease morbidity associates with and may in part be due to epithelial barrier leak. An equally large body of published literature substantiates that a diverse group of micronutrients can reduce barrier leak across a wide array of epithelial tissue types, stemming from both cell culture as well as animal and human tissue models. Conversely, micronutrient deficiencies can exacerbate both barrier leak and morbidity. Focusing on zinc, Vitamin A and Vitamin D, this review shows that at concentrations above RDA levels but well below toxicity limits, these micronutrients can induce cell- and tissue-specific molecular-level changes in tight junctional complexes (and by other mechanisms) that reduce barrier leak. An opportunity now exists in critical care—but also medical prophylactic and therapeutic care in general—to consider implementation of select micronutrients at elevated dosages as adjuvant therapeutics in a variety of disease management. This consideration is particularly pointed amidst the COVID-19 pandemic.
Collapse
|
49
|
Riedel S, Pheiffer C, Johnson R, Louw J, Muller CJF. Intestinal Barrier Function and Immune Homeostasis Are Missing Links in Obesity and Type 2 Diabetes Development. Front Endocrinol (Lausanne) 2022; 12:833544. [PMID: 35145486 PMCID: PMC8821109 DOI: 10.3389/fendo.2021.833544] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Noncommunicable diseases, such as type 2 diabetes (T2D), place a burden on healthcare systems worldwide. The rising prevalence of obesity, a major risk factor for T2D, is mainly attributed to the adoption of Westernized diets and lifestyle, which cause metabolic dysfunction and insulin resistance. Moreover, diet may also induce changes in the microbiota composition, thereby affecting intestinal immunity. The critical role of intestinal immunity and intestinal barrier function in the development of T2D is increasingly acknowledged, however, limited studies have investigated the link between intestinal function and metabolic disease. In this review, studies reporting specific roles of the intestinal immune system and intestinal epithelial cells (IECs) in metabolic disease are highlighted. Innate chemokine signaling, eosinophils, immunoglobulin A (IgA), T helper (Th) 17 cells and their cytokines were associated with obesity and/or dysregulated glucose homeostasis. Intestinal epithelial cells (IECs) emerged as critical modulators of obesity and glucose homeostasis through their effect on lipopolysaccharide (LPS) signaling and decontamination. Furthermore, IECs create a link between microbial metabolites and whole-body metabolic function. Future in depth studies of the intestinal immune system and IECs may provide new opportunities and targets to develop treatments and prevention strategies for obesity and T2D.
Collapse
Affiliation(s)
- Sylvia Riedel
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
- Department of Obstetrics and Gynaecology, University of Pretoria, Pretoria, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Christo J. F. Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| |
Collapse
|
50
|
Suda K, Matsuda K. How Microbes Affect Depression: Underlying Mechanisms via the Gut-Brain Axis and the Modulating Role of Probiotics. Int J Mol Sci 2022; 23:ijms23031172. [PMID: 35163104 PMCID: PMC8835211 DOI: 10.3390/ijms23031172] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Accumulating evidence suggests that the gut microbiome influences the brain functions and psychological state of its host via the gut-brain axis, and gut dysbiosis has been linked to several mental illnesses, including major depressive disorder (MDD). Animal experiments have shown that a depletion of the gut microbiota leads to behavioral changes, and is associated with pathological changes, including abnormal stress response and impaired adult neurogenesis. Short-chain fatty acids such as butyrate are known to contribute to the up-regulation of brain-derived neurotrophic factor (BDNF), and gut dysbiosis causes decreased levels of BDNF, which could affect neuronal development and synaptic plasticity. Increased gut permeability causes an influx of gut microbial components such as lipopolysaccharides, and the resultant systemic inflammation may lead to neuroinflammation in the central nervous system. In light of the fact that gut microbial factors contribute to the initiation and exacerbation of depressive symptoms, this review summarizes the current understanding of the molecular mechanisms involved in MDD onset, and discusses the therapeutic potential of probiotics, including butyrate-producing bacteria, which can mediate the microbiota-gut-brain axis.
Collapse
|