1
|
Patloka O, Komprda T, Franke G. Review of the Relationships Between Human Gut Microbiome, Diet, and Obesity. Nutrients 2024; 16:3996. [PMID: 39683390 DOI: 10.3390/nu16233996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity is a complex disease that increases the risk of other pathologies. Its prevention and long-term weight loss maintenance are problematic. Gut microbiome is considered a potential obesity modulator. The objective of the present study was to summarize recent findings regarding the relationships between obesity, gut microbiota, and diet (vegetable/animal proteins, high-fat diets, restriction of carbohydrates), with an emphasis on dietary fiber and resistant starch. The composition of the human gut microbiome and the methods of its quantification are described. Products of the gut microbiome metabolism, such as short-chain fatty acids and secondary bile acids, and their effects on the gut microbiota, intestinal barrier function and immune homeostasis are discussed in the context of obesity. The importance of dietary fiber and resistant starch is emphasized as far as effects of the host diet on the composition and function of the gut microbiome are concerned. The complex relationships between human gut microbiome and obesity are finally summarized.
Collapse
Affiliation(s)
- Ondřej Patloka
- Department of Food Technology, Mendel University in Brno, 61300 Brno, Czech Republic
| | - Tomáš Komprda
- Department of Food Technology, Mendel University in Brno, 61300 Brno, Czech Republic
| | - Gabriela Franke
- Department of Food Technology, Mendel University in Brno, 61300 Brno, Czech Republic
| |
Collapse
|
2
|
Wu Q, Li P, Li X, Ma L, Chen K, Man S. Pueraria Extract Ameliorates Alcoholic Liver Disease via the Liver-Gut-Brain Axis: Focus on Restoring the Intestinal Barrier and Inhibiting Alcohol Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24449-24462. [PMID: 39445550 DOI: 10.1021/acs.jafc.4c05365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Alcoholic liver disease (ALD) is one of the causes of hepatocellular carcinoma, accompanied by intestinal leakage and microbial changes. Pueraria has protective effects on liver injury. The aim of this study was to investigate the mechanism of pueraria in the treatment of ALD. UPLC-Q/TOF-MS was used to analyze the composition of the pueraria extract (PUE). Acute and chronic ALD models were established to evaluate the antialcoholic and hepatoprotective effects of PUE. As a result, PUE treatment reduced the serum levels of ALT, AST, TC, and TG and inflammatory factors and alleviated liver inflammation and drunk state. PUE decreased the gene expression of ADH1 and the serum level of acetaldehyde (ACH) to inhibit the generation of ACH from ethanol metabolism, increased the gene level of ALDH2 to accelerate the decomposition of ACH, and thereby alleviated liver inflammation and intestinal barrier damage. Meanwhile, 16 S rDNA revealed that PUE altered the microbiota composition, reduced the amount of Proteobacteria and Desulfobacterota, and thus inhibited the generation of lipopolysaccharide and its downstream-like TLR4/MyD88/NF-κB pathway. PUE also increased the abundance of Bacteroides, Ruminococcus, and Prevotella and producted short-chain fatty acids to protect the intestinal wall. Treatment with fecal microbiota transplantation further confirmed that PUE gut microbiota dependently alleviated ALD. Therefore, PUE regulated gut microbiota and inhibited ethanol metabolism to alleviate ALD through the liver-gut-brain axis. It has good prospects in the future.
Collapse
Affiliation(s)
- Qiong Wu
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Piaoran Li
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Xuejiao Li
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Kai Chen
- The Innovation Institute of Agricultural Technology, College of Life Science, Shangrao Normal University, Shangrao 334001, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| |
Collapse
|
3
|
Saenz E, Montagut NE, Wang B, Stein-Thöringer C, Wang K, Weng H, Ebert M, Schneider KM, Li L, Teufel A. Manipulating the Gut Microbiome to Alleviate Steatotic Liver Disease: Current Progress and Challenges. ENGINEERING 2024; 40:51-60. [DOI: 10.1016/j.eng.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Bock PM, Martins AF, Schaan BD. Understanding how pre- and probiotics affect the gut microbiome and metabolic health. Am J Physiol Endocrinol Metab 2024; 327:E89-E102. [PMID: 38809510 DOI: 10.1152/ajpendo.00054.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
The gut microbiome, a complex assembly of microorganisms, significantly impacts human health by influencing nutrient absorption, the immune system, and disease response. These microorganisms form a dynamic ecosystem that is critical to maintaining overall well-being. Prebiotics and probiotics are pivotal in regulating gut microbiota composition. Prebiotics nourish beneficial bacteria and promote their growth, whereas probiotics help maintain balance within the microbiome. This intricate balance extends to several aspects of health, including maintaining the integrity of the gut barrier, regulating immune responses, and producing metabolites crucial for metabolic health. Dysbiosis, or an imbalance in the gut microbiota, has been linked to metabolic disorders such as type 2 diabetes, obesity, and cardiovascular disease. Impaired gut barrier function, endotoxemia, and low-grade inflammation are associated with toll-like receptors influencing proinflammatory pathways. Short-chain fatty acids derived from microbial fermentation modulate anti-inflammatory and immune system pathways. Prebiotics positively influence gut microbiota, whereas probiotics, especially Lactobacillus and Bifidobacterium strains, may improve metabolic outcomes, such as glycemic control in diabetes. It is important to consider strain-specific effects and study variability when interpreting these findings, highlighting the need for further research to optimize their therapeutic potential. The aim of this report is therefore to review the role of the gut microbiota in metabolic health and disease and the effects of prebiotics and probiotics on the gut microbiome and their therapeutic role, integrating a broad understanding of physiological mechanisms with a clinical perspective.
Collapse
Affiliation(s)
- Patricia M Bock
- Pharmacology, Institute of Basic Science, Universidade Federal do Rio Grande, Rio Grande, Brazil
| | - Andreza F Martins
- Microbiology, Department of Microbiology, Immunology, and Parasitology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Beatriz D Schaan
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
5
|
Valladares L, Vio Del Río F. [Bioactive components of whole grain and their effect on health]. NUTR HOSP 2024; 41:706-711. [PMID: 38501834 DOI: 10.20960/nh.04986] [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] [Indexed: 03/20/2024] Open
Abstract
Introduction Non-communicable diseases represent a global health burden with increasing prevalence. To prevent or improve this type of diseases, dietary strategies based on healthy foods have been suggested. Cereals are the most consumed foods in the world and preventive effects of whole grains on health have been described. The germ and bran of cereals are abundant in bioactive compounds, including phytochemicals, vitamins, minerals and fibers, and these compounds are effective in preventing and improving non-communicable diseases. This review analyzes the content and distribution of the primary components of whole grains (wheat, barley, oats, rice and black wheat) and their fractions, focusing on the mechanisms by which phenolic acids and dietary fiber contribute to reducing the risk of metabolic and cardiovascular diseases and cancer. There is clear evidence of the broad cellular and physiological effects of bioactive compounds in whole grains, supporting the health value of a diet rich in whole grains.
Collapse
Affiliation(s)
- Luis Valladares
- Instituto de Nutrición y Tecnología de los Alimentos (INTA). Universidad de Chile
| | - Fernando Vio Del Río
- Instituto de Nutrición y Tecnología de los Alimentos (INTA). Universidad de Chile
| |
Collapse
|
6
|
Puhlmann ML, van de Rakt E, Kerezoudi EN, Rangel I, Brummer RJ, Smidt H, Kaper FS, de Vos WM. Analysis of the fermentation kinetics and gut microbiota modulatory effect of dried chicory root reveals the impact of the plant-cell matrix rationalizing its conversion in the distal colon. MICROBIOME RESEARCH REPORTS 2024; 3:28. [PMID: 39421250 PMCID: PMC11485554 DOI: 10.20517/mrr.2024.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/04/2024] [Accepted: 04/19/2024] [Indexed: 10/19/2024]
Abstract
Aim: The cell matrix of plant foods has received little attention in prebiotic fiber research. We aimed to understand the impact of the plant cell matrix in dried chicory root on its breakdown in the human gut to explain its reported beneficial effects on gut and metabolic health. Methods: We applied in vitro digestion and fermentation models together with an ex vivo gut barrier integrity model. Plant cell matrix intactness in the upper gastrointestinal tract was investigated by scanning electron microscopy. Colonic breakdown of inulin, and chicory root cubes and powder was assessed by gut microbiota analysis using 16S rRNA gene amplicon sequencing and determining the kinetics of changes in pH, gas, and short-chain fatty acid (SCFA) production. Finally, effects on gut barrier integrity were explored by exposing colonic biopsies to fermentation supernatants in an Ussing chamber model. Results: The plant cell matrix of dried chicory root cubes remained intact throughout upper gastrointestinal transit. Dried chicory root fermentation resulted in higher final relative abundances of pectin-degrading Monoglobus and butyrate-producing Roseburia spp. compared to inulin and a seven-fold increase in Bifidobacterium spp. in donors where these species were present. Dried chicory root cubes yielded similar total SCFAs but higher final butyrate levels than chicory root powder or isolated inulin with less gas produced. No uniform but donor-specific effects of fermentation supernatants on the maintenance of gut barrier integrity were detected. Conclusion: The intact plant cell matrix of dried chicory root affected its colonic breakdown kinetics and microbiota, underpinning its beneficial effect in vivo.
Collapse
Affiliation(s)
- Marie-Luise Puhlmann
- Laboratory of Microbiology, Wageningen University & Research, Wageningen 6708 WE, the Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen 6708 WE, the Netherlands
| | - Ember van de Rakt
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen 6708 WE, the Netherlands
| | - Evangelia N. Kerezoudi
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro 70182, Sweden
- Department of Nutrition and Dietetics, Harokopio University, Athens 17671, Greece
| | - Ignacio Rangel
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro 70182, Sweden
| | - Robert J. Brummer
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro 70182, Sweden
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen 6708 WE, the Netherlands
| | | | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University & Research, Wageningen 6708 WE, the Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland
| |
Collapse
|
7
|
Donkers JM, Wiese M, van den Broek TJ, Wierenga E, Agamennone V, Schuren F, van de Steeg E. A host-microbial metabolite interaction gut-on-a-chip model of the adult human intestine demonstrates beneficial effects upon inulin treatment of gut microbiome. MICROBIOME RESEARCH REPORTS 2024; 3:18. [PMID: 38841408 PMCID: PMC11149092 DOI: 10.20517/mrr.2023.79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/29/2024] [Accepted: 02/20/2024] [Indexed: 06/07/2024]
Abstract
Background: The gut and its microbiome have a major impact on many aspects of health and are therefore also an attractive target for drug- or food-based therapies. Here, we report on the added value of combining a microbiome screening model, the i-screen, with fresh intestinal tissue explants in a microfluidic gut-on-a-chip model, the Intestinal Explant Barrier Chip (IEBC). Methods: Adult human gut microbiome (fecal pool of 6 healthy donors) was cultured anaerobically in the i-screen platform for 24 h, without and with exposure to 4 mg/mL inulin. The i-screen cell-free culture supernatant was subsequently applied to the luminal side of adult human colon tissue explants (n = 3 donors), fixed in the IEBC, for 24 h and effects were evaluated. Results: The supplementation of the media with inulin promoted the growth of Anaerostipes, Bifidobacterium, Blautia, and Collinsella in the in vitro i-screen, and triggered an elevated production of butyrate by the microbiota. Human colon tissue exposed to inulin-treated i-screen cell-free culture supernatant or control i-screen cell-free culture supernatant with added short-chain fatty acids (SCFAs) showed improved tissue barrier integrity measured by a 28.2%-34.2% reduction in FITC-dextran 4000 (FD4) leakage and 1.3 times lower transport of antipyrine. Furthermore, the release of pro-inflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α was reduced under these circumstances. Gene expression profiles confirmed these findings, but showed more profound effects for inulin-treated supernatant compared to SCFA-supplemented supernatant. Conclusion: The combination of i-screen and IEBC facilitates the study of complex intestinal processes such as host-microbial metabolite interaction and gut health.
Collapse
Affiliation(s)
- Joanne M. Donkers
- Department of Metabolic Health Research, TNO, Leiden 2333 BE, the Netherlands
| | - Maria Wiese
- Department of Microbiology & Systems Biology, TNO, Leiden 2333 BE, the Netherlands
| | - Tim J. van den Broek
- Department of Microbiology & Systems Biology, TNO, Leiden 2333 BE, the Netherlands
| | - Esmée Wierenga
- Department of Metabolic Health Research, TNO, Leiden 2333 BE, the Netherlands
| | - Valeria Agamennone
- Department of Microbiology & Systems Biology, TNO, Leiden 2333 BE, the Netherlands
| | - Frank Schuren
- Department of Microbiology & Systems Biology, TNO, Leiden 2333 BE, the Netherlands
| | - Evita van de Steeg
- Department of Metabolic Health Research, TNO, Leiden 2333 BE, the Netherlands
| |
Collapse
|
8
|
Molotla-Torres DE, Guzmán-Mejía F, Godínez-Victoria M, Drago-Serrano ME. Role of Stress on Driving the Intestinal Paracellular Permeability. Curr Issues Mol Biol 2023; 45:9284-9305. [PMID: 37998758 PMCID: PMC10670774 DOI: 10.3390/cimb45110581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/05/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
The gut epithelium is a polarized monolayer that exhibits apical and basolateral membrane surfaces. Monolayer cell components are joined side by side via protein complexes known as tight junction proteins (TJPs), expressed at the most apical extreme of the basolateral membrane. The gut epithelium is a physical barrier that determinates intestinal permeability, referred to as the measurement of the transit of molecules from the intestinal lumen to the bloodstream or, conversely, from the blood to the gut lumen. TJPs play a role in the control of intestinal permeability that can be disrupted by stress through signal pathways triggered by the ligation of receptors with stress hormones like glucocorticoids. Preclinical studies conducted under in vitro and/or in vivo conditions have addressed underlying mechanisms that account for the impact of stress on gut permeability. These mechanisms may provide insights for novel therapeutic interventions in diseases in which stress is a risk factor, like irritable bowel syndrome. The focus of this study was to review, in an integrative context, the neuroendocrine effects of stress, with special emphasis on TJPs along with intestinal permeability.
Collapse
Affiliation(s)
- Daniel Efrain Molotla-Torres
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Calzada del Hueso No. 1100, Ciudad de México CP 04960, Mexico;
| | - Fabiola Guzmán-Mejía
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Calzada del Hueso No. 1100, Ciudad de México CP 04960, Mexico
| | - Marycarmen Godínez-Victoria
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Ciudad de México CP 11340, Mexico;
| | - Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Calzada del Hueso No. 1100, Ciudad de México CP 04960, Mexico
| |
Collapse
|
9
|
Almeida C, Gonçalves-Nobre JG, Alpuim Costa D, Barata P. The potential links between human gut microbiota and cardiovascular health and disease - is there a gut-cardiovascular axis? FRONTIERS IN GASTROENTEROLOGY 2023; 2. [DOI: 10.3389/fgstr.2023.1235126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The gut-heart axis is an emerging concept highlighting the crucial link between gut microbiota and cardiovascular diseases (CVDs). Recent studies have demonstrated that gut microbiota is pivotal in regulating host metabolism, inflammation, and immune function, critical drivers of CVD pathophysiology. Despite a strong link between gut microbiota and CVDs, this ecosystem’s complexity still needs to be fully understood. The short-chain fatty acids, trimethylamine N-oxide, bile acids, and polyamines are directly or indirectly involved in the development and prognosis of CVDs. This review explores the relationship between gut microbiota metabolites and CVDs, focusing on atherosclerosis and hypertension, and analyzes personalized microbiota-based modulation interventions, such as physical activity, diet, probiotics, prebiotics, and fecal microbiota transplantation, as a promising strategy for CVD prevention and treatment.
Collapse
|
10
|
Wang K, Wang K, Wang J, Yu F, Ye C. Protective Effect of Clostridium butyricum on Escherichia coli-Induced Endometritis in Mice via Ameliorating Endometrial Barrier and Inhibiting Inflammatory Response. Microbiol Spectr 2022; 10:e0328622. [PMID: 36321897 PMCID: PMC9769554 DOI: 10.1128/spectrum.03286-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022] Open
Abstract
Endometritis is a common reproductive disease occurs both in human and animals. Clostridium butyricum is a Gram-positive anaerobic bacterium that can ferment various carbohydrates into butyric acid. In this study, we investigated the effects of C. butyricum on Escherichia coli-induced endometritis and clarified the underlying mechanism. We first verified the protective effect of C. butyricum in vivo by establishing a mouse model of E. coli-induced endometritis. It was determined that C. butyricum pretreatment significantly reversed E. coli-induced uterine histopathological changes. Meanwhile, C. butyricum pretreatment significantly decreased the production of pro-inflammatory mediators and the levels of myeloperoxidase (MPO) and malondialdehyde (MDA). We found that C. butyricum could inhibit TLR4-mediated phosphorylation of NF-κB and the activity of histone deacetylase (HDAC). Furthermore, C. butyricum significantly increased the expression of the tight junction proteins (TJPs) ZO-1, claudin-3, and occludin. Additionally, treatment with C. butyricum culture supernatant dramatically suppressed the degree of inflammation in the uterus, and inactivated C. butyricum did not exert a protective effect. We subsequently investigated butyrate levels in both the uterus and blood and observed a marked augment in the C. butyricum treatment group. Collectively, our data suggest that C. butyricum maintains epithelial barrier function and suppresses inflammatory response during E. coli-induced endometritis and that the protective effect of C. butyricum may be related to the production of butyrate. IMPORTANCE Endometritis is a common reproductive disease both in human and animals. It impairs female fertility by disrupting endometrial function. Antibiotics are widely used to treat endometritis in clinical practice, but the misuse of antibiotics often leads to antibiotic resistance. Therefore, there is an urgent need for new therapeutic agents to treat bacterial endometritis and overcome bacterial resistance. In this study, we found that C. butyricum could protect from E. coli-induced endometritis.
Collapse
Affiliation(s)
- Kexin Wang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Ke Wang
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Junrong Wang
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Fan Yu
- Department of Gastroenterology and Hepatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Cong Ye
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
11
|
Aczel D, Gyorgy B, Bakonyi P, BukhAri R, Pinho R, Boldogh I, Yaodong G, Radak Z. The Systemic Effects of Exercise on the Systemic Effects of Alzheimer's Disease. Antioxidants (Basel) 2022; 11:antiox11051028. [PMID: 35624892 PMCID: PMC9137920 DOI: 10.3390/antiox11051028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive degenerative disorder and a leading cause of dementia in the elderly. The etiology of AD is multifactorial, including an increased oxidative state, deposition of amyloid plaques, and neurofibrillary tangles of the tau protein. The formation of amyloid plaques is considered one of the first signs of the illness, but only in the central nervous system (CNS). Interestingly, results indicate that AD is not just localized in the brain but is also found in organs distant from the brain, such as the cardiovascular system, gut microbiome, liver, testes, and kidney. These observations make AD a complex systemic disorder. Still, no effective medications have been found, but regular physical activity has been considered to have a positive impact on this challenging disease. While several articles have been published on the benefits of physical activity on AD development in the CNS, its peripheral effects have not been discussed in detail. The provocative question arising is the following: is it possible that the beneficial effects of regular exercise on AD are due to the systemic impact of training, rather than just the effects of exercise on the brain? If so, does this mean that the level of fitness of these peripheral organs can directly or indirectly influence the incidence or progress of AD? Therefore, the present paper aims to summarize the systemic effects of both regular exercise and AD and point out how common exercise-induced adaptation via peripheral organs can decrease the incidence of AD or attenuate the progress of AD.
Collapse
Affiliation(s)
- Dora Aczel
- Research Institute of Sport Science, University of Physical Education, 1123 Budapest, Hungary; (D.A.); (B.G.); (P.B.); (R.B.)
| | - Bernadett Gyorgy
- Research Institute of Sport Science, University of Physical Education, 1123 Budapest, Hungary; (D.A.); (B.G.); (P.B.); (R.B.)
| | - Peter Bakonyi
- Research Institute of Sport Science, University of Physical Education, 1123 Budapest, Hungary; (D.A.); (B.G.); (P.B.); (R.B.)
| | - RehAn BukhAri
- Research Institute of Sport Science, University of Physical Education, 1123 Budapest, Hungary; (D.A.); (B.G.); (P.B.); (R.B.)
| | - Ricardo Pinho
- Laboratory of Exercise Biochemistry in Health, Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil;
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA;
| | - Gu Yaodong
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China;
| | - Zsolt Radak
- Research Institute of Sport Science, University of Physical Education, 1123 Budapest, Hungary; (D.A.); (B.G.); (P.B.); (R.B.)
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
- Correspondence: ; Tel.: +36-1-3565764; Fax: +36-1-3566337
| |
Collapse
|
12
|
Kramer P. Mitochondria-Microbiota Interaction in Neurodegeneration. Front Aging Neurosci 2022; 13:776936. [PMID: 35002678 PMCID: PMC8733591 DOI: 10.3389/fnagi.2021.776936] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s and Parkinson’s are the two best-known neurodegenerative diseases. Each is associated with the excessive aggregation in the brain and elsewhere of its own characteristic amyloid proteins. Yet the two afflictions have much in common and often the same amyloids play a role in both. These amyloids need not be toxic and can help regulate bile secretion, synaptic plasticity, and immune defense. Moreover, when they do form toxic aggregates, amyloids typically harm not just patients but their pathogens too. A major port of entry for pathogens is the gut. Keeping the gut’s microbe community (microbiota) healthy and under control requires that our cells’ main energy producers (mitochondria) support the gut-blood barrier and immune system. As we age, these mitochondria eventually succumb to the corrosive byproducts they themselves release, our defenses break down, pathogens or their toxins break through, and the side effects of inflammation and amyloid aggregation become problematic. Although it gets most of the attention, local amyloid aggregation in the brain merely points to a bigger problem: the systemic breakdown of the entire human superorganism, exemplified by an interaction turning bad between mitochondria and microbiota.
Collapse
Affiliation(s)
- Peter Kramer
- Department of General Psychology, University of Padua, Padua, Italy
| |
Collapse
|
13
|
Olsthoorn L, Vreeken D, Kiliaan AJ. Gut Microbiome, Inflammation, and Cerebrovascular Function: Link Between Obesity and Cognition. Front Neurosci 2021; 15:761456. [PMID: 34938153 PMCID: PMC8685335 DOI: 10.3389/fnins.2021.761456] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/16/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity affects 13% of the adult population worldwide and this number is only expected to increase. Obesity is known to have a negative impact on cardiovascular and metabolic health, but it also impacts brain structure and function; it is associated with both gray and white matter integrity loss, as well as decreased cognitive function, including the domains of executive function, memory, inhibition, and language. Especially midlife obesity is associated with both cognitive impairment and an increased risk of developing dementia at later age. However, underlying mechanisms are not yet fully revealed. Here, we review recent literature (published between 2010 and March 2021) and discuss the effects of obesity on brain structure and cognition, with a main focus on the contributions of the gut microbiome, white adipose tissue (WAT), inflammation, and cerebrovascular function. Obesity-associated changes in gut microbiota composition may cause increased gut permeability and inflammation, therewith affecting cognitive function. Moreover, excess of WAT in obesity produces pro-inflammatory adipokines, leading to a low grade systemic peripheral inflammation, which is associated with decreased cognition. The blood-brain barrier also shows increased permeability, allowing among others, peripheral pro-inflammatory markers to access the brain, leading to neuroinflammation, especially in the hypothalamus, hippocampus and amygdala. Altogether, the interaction between the gut microbiota, WAT inflammation, and cerebrovascular integrity plays a significant role in the link between obesity and cognition. Future research should focus more on the interplay between gut microbiota, WAT, inflammation and cerebrovascular function to obtain a better understanding about the complex link between obesity and cognitive function in order to develop preventatives and personalized treatments.
Collapse
Affiliation(s)
- Lisette Olsthoorn
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Netherlands
| | - Debby Vreeken
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Netherlands.,Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, Arnhem, Netherlands
| | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Netherlands
| |
Collapse
|
14
|
Cookson TA. Bacterial-Induced Blood Pressure Reduction: Mechanisms for the Treatment of Hypertension via the Gut. Front Cardiovasc Med 2021; 8:721393. [PMID: 34485420 PMCID: PMC8414577 DOI: 10.3389/fcvm.2021.721393] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/16/2021] [Indexed: 01/08/2023] Open
Abstract
Hypertension is a major risk factor for the development of cardiovascular disease. As more research into the gut microbiome emerges, we are finding increasing evidence to support that these microbes may have significant positive and negative effects on blood pressure and associated disorders. The bacterial-derived metabolites that are produced in the gut are capable of widespread effects to several tissue types and organs in the body. It is clear that the extensive metabolic function that is lost with gut dysbiosis is unlikely to be replenished with a single metabolite or bacterial strain. Instead, combinations of bacteria and concomitant therapies will provide a more well-rounded solution to manage hypertension. The bioactive molecules that are recognized in this review will inform on ideal characteristics of candidate bacteria and provide direction for future research on the gut microbiome in hypertension.
Collapse
|
15
|
Hu Q, Yin F, Li B, Guo Y, Yin Y. Dietary Tributyrin Administration Improves Intestinal Morphology and Selected Bacterial and Short-Chain Fatty Acid Profiles in Broilers Under an Isocaloric Feeding Regime. Front Microbiol 2021; 12:715712. [PMID: 34421875 PMCID: PMC8371336 DOI: 10.3389/fmicb.2021.715712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/12/2021] [Indexed: 12/17/2022] Open
Abstract
The current study was conducted to investigate the effect of dietary tributyrin (TB) administration on the intestinal and growth performances in Arbor Acres (AA) broilers under an isocaloric feeding regime. A total of 540 day-old healthy AA broilers were randomly assigned to five treatments with 12 replicates (pens) per treatment and nine birds per pen for 42 days. The dietary treatments were basal diet (control) and basal diet with TB at doses of 0.23 g/kg (TB1), 0.46 g/kg (TB2), 0.92 g/kg (TB3), and 1.84 g/kg (TB4). Particularly, to achieve the isocaloric and cost-saving experimental diets, soybean oil was replaced by the TB product (Eucalorie®) with equivalent metabolic energy contents, and the formulas were rebalanced with zeolite to get the sum of all the feed ingredients to 100%. On days 21 and 42, after weighing, the birds (one bird per replicate) whose body weight was close to the replicate average were euthanized to investigate the effect of dietary TB on intestinal morphology, intestinal bacterial population, and short-chain fatty acid contents. The results revealed that dietary TB administration increased the average daily gain, gain/feed ratio, and European broiler index (P < 0.05) and improved the intestinal morphology (P < 0.05) as indicated by higher villus height and the ratios of villus height/crypt depth in broilers. The incremental levels of TB increased the ileal Lactobacillus content (P = 0.05) and cecal Bacillus content (P = 0.02), respectively. Moreover, dietary TB administration also increased the contents of most of the selected short-chain fatty acids in ileal and cecal digesta (P < 0.05). Collectively, dietary TB administration quadratically improved the growth performance, intestinal morphology, beneficial bacterial population, and short-chain fatty acid levels under the isocaloric feeding regime, indicating better profit return potential in practical poultry operation.
Collapse
Affiliation(s)
- Qunbing Hu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,Hubei Horwath Biotechnology Co., Ltd., Xianning, China
| | - Fugui Yin
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,Hubei Horwath Biotechnology Co., Ltd., Xianning, China
| | - Baocheng Li
- Hubei Horwath Biotechnology Co., Ltd., Xianning, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yulong Yin
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
16
|
Jones-Freeman B, Chonwerawong M, Marcelino VR, Deshpande AV, Forster SC, Starkey MR. The microbiome and host mucosal interactions in urinary tract diseases. Mucosal Immunol 2021; 14:779-792. [PMID: 33542492 DOI: 10.1038/s41385-020-00372-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023]
Abstract
The urinary tract consists of the bladder, ureters, and kidneys, and is an essential organ system for filtration and excretion of waste products and maintaining systemic homeostasis. In this capacity, the urinary tract is impacted by its interactions with other mucosal sites, including the genitourinary and gastrointestinal systems. Each of these sites harbors diverse ecosystems of microbes termed the microbiota, that regulates complex interactions with the local and systemic immune system. It remains unclear whether changes in the microbiota and associated metabolites may be a consequence or a driver of urinary tract diseases. Here, we review the current literature, investigating the impact of the microbiota on the urinary tract in homeostasis and disease including urinary stones, acute kidney injury, chronic kidney disease, and urinary tract infection. We propose new avenues for exploration of the urinary microbiome using emerging technology and discuss the potential of microbiome-based medicine for urinary tract conditions.
Collapse
Affiliation(s)
- Bernadette Jones-Freeman
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Michelle Chonwerawong
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Vanessa R Marcelino
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Aniruddh V Deshpande
- Priority Research Centre GrowUpWell, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia.,Department of Pediatric Urology and Surgery, John Hunter Children's Hospital, New Lambton Heights, NSW, Australia.,Urology Unit, Department of Pediatric Surgery, Children's Hospital at Westmead, Sydney Children's Hospital Network, Westmead, NSW, Australia
| | - Samuel C Forster
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Malcolm R Starkey
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia. .,Priority Research Centre GrowUpWell, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|
17
|
Yun Y, Baek A, Kim DE. Autophagy down-regulates NLRP3-dependent inflammatory response of intestinal epithelial cells under nutrient deprivation. BMB Rep 2021. [PMID: 33407996 PMCID: PMC8167250 DOI: 10.5483/bmbrep.2021.54.5.211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dysregulation of inflammation induced by noninfectious stress conditions, such as nutrient deprivation, causes tissue damage and intestinal permeability, resulting in the development of inflammatory bowel diseases. We studied the effect of autophagy on cytokine secretion related to intestinal permeability under nutrient deprivation. Autophagy removes NLRP3 inflammasomes via ubiquitin-mediated degradation under starvation. When autophagy was inhibited, starvation-induced NLRP3 inflammasomes and their product, IL-1β, were significantly enhanced. A prolonged nutrient deprivation resulted in an increased epithelial mesenchymal transition (EMT), leading to intestinal permeability. Under nutrient deprivation, IL-17E/25, which is secreted by IL-1β, demolished the intestinal epithelial barrier. Our results suggest that an upregulation of autophagy maintains the intestinal barrier by suppressing the activation of NLRP3 inflammasomes and the release of their products, including proinflammatory cytokines IL-1β and IL-17E/25, under nutrient deprivation.
Collapse
Affiliation(s)
- Yewon Yun
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Ahruem Baek
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
18
|
Zhang Z, Zhang Q, Li F, Xin Y, Duan Z. Contributions of HO-1-Dependent MAPK to Regulating Intestinal Barrier Disruption. Biomol Ther (Seoul) 2021; 29:175-183. [PMID: 33093265 PMCID: PMC7921856 DOI: 10.4062/biomolther.2020.112] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/08/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022] Open
Abstract
The mitogen-activated protein kinase (MAPK) pathway controls intestinal epithelial barrier permeability by regulating tight junctions (TJs) and epithelial cells damage. Heme oxygenase-1 (HO-1) and carbon monoxide (CO) protect the intestinal epithelial barrier function, but the molecular mechanism is not yet clarified. MAPK activation and barrier permeability were studied using monolayers of Caco-2 cells treated with tissue necrosis factor α (TNF-α) transfected with FUGW-HO-1 or pLKO.1-sh-HO-1 plasmid. Intestinal mucosal barrier permeability and MAPK activation were also investigated using carbon tetrachloride (CCl4) administration with CoPP (a HO-1 inducer), ZnPP (a HO-1 inhibitor), CO releasing molecule 2 (CORM-2), or inactived-CORM-2-treated wild-type mice and mice with HO-1 deficiency in intestinal epithelial cells. TNF-α increased epithelial TJ disruption and cleaved caspase-3 expression, induced ERK, p38, and JNK phosphorylation. In addition, HO-1 blocked TNF-α-induced increase in epithelial TJs disruption, cleaved caspase-3 expression, as well as ERK, p38, and JNK phosphorylation in an HO-1-dependent manner. CoPP and CORM-2 directly ameliorated intestinal mucosal injury, attenuated TJ disruption and cleaved caspase-3 expression, and inhibited epithelial ERK, p38, and JNK phosphorylation after chronic CCl4 injection. Conversely, ZnPP completely reversed these effects. Furthermore, mice with intestinal epithelial HO-1 deficient exhibited a robust increase in mucosal TJs disruption, cleaved caspase-3 expression, and MAPKs activation as compared to the control group mice. These data demonstrated that HO-1-dependent MAPK signaling inhibition preserves the intestinal mucosal barrier integrity by abrogating TJ dysregulation and epithelial cell damage. The differential targeting of gut HO-1-MAPK axis leads to improved intestinal disease therapy.
Collapse
Affiliation(s)
- Zhenling Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian116011, China
| | - Qiuping Zhang
- Department of Pathology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Fang Li
- Department of Immunology, Dalian Medical University, Dalian 116044, China
| | - Yi Xin
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China
| | - Zhijun Duan
- Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian116011, China
| |
Collapse
|