1
|
Zhao X, Zhu M, Wang Z, Gao M, Long Y, Zhou S, Wang W. The Alleviative Effect of Sodium Butyrate on Dexamethasone-Induced Skeletal Muscle Atrophy. Cell Biol Int 2025; 49:508-521. [PMID: 39936899 DOI: 10.1002/cbin.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 01/18/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Skeletal muscle mass is significantly negatively regulated by glucocorticoids. Following glucocorticoid administration, the balance between protein synthesis and breakdown in skeletal muscle is disrupted, shifting towards a predominance of catabolic metabolism. Short-chain fatty acids like sodium butyrate have been found to regulate inflammatory reactions and successively activate signaling pathways. The preventive benefits of sodium butyrate against dexamethasone-induced skeletal muscle atrophy and myotube atrophy models were examined in this work, and the underlying mechanism was clarified. A total of 32 6-week-old C57BL/6 inbred male mice were randomly assigned to one of four groups and treated with dexamethasone to induce muscle atrophy and sodium butyrate. We found that sodium succinate alleviated dexamethasone-induced myotube atrophy in the myotube atrophy model by lowering the gene expression of two E3 ubiquitin ligases, Atrogin-1 and MURF1, and activating the AKT/mTOR signaling pathway. Pertussis toxin reversed this effect, indicating that G protein-coupled receptors were involved in sodium butyrate's action as a mediator. Additionally, pre-treatment with sodium butyrate lowered weight and muscle mass loss in a mouse model of skeletal muscle atrophy, dramatically decreased the MURF1 gene expression and decreased the nuclear translocation of the glucocorticoid receptor. In conclusion, this study shows that sodium butyrate inhibits the expression of atrophy genes, thus preventing the breakdown of proteins and the loss of muscle mass, while also inhibiting weight loss, in animal models.
Collapse
Affiliation(s)
- Xingchen Zhao
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Mingqiang Zhu
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Zifan Wang
- College of Animal Science and Veterinary, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Ming Gao
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Yifei Long
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Shuo Zhou
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Wei Wang
- Innovative Institute of Animal Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
- College of Animal Science and Veterinary, Shenyang Agricultural University, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Ho IHT, Zou Y, Luo K, Qin F, Jiang Y, Li Q, Jin T, Zhang X, Chen H, Tan L, Zhang L, Gin T, Wu WKK, Chan MTV, Jiang C, Liu X. Sodium butyrate restored TRESK current controlling neuronal hyperexcitability in a mouse model of oxaliplatin-induced peripheral neuropathic pain. Neurotherapeutics 2025; 22:e00481. [PMID: 39542827 PMCID: PMC11742850 DOI: 10.1016/j.neurot.2024.e00481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/30/2024] [Accepted: 10/27/2024] [Indexed: 11/17/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) and its related pain are common challenges for patients receiving oxaliplatin chemotherapy. Oxaliplatin accumulation in dorsal root ganglion (DRGs) is known to impair gene transcription by epigenetic dysregulation. We hypothesized that sodium butyrate, a pro-resolution short-chain fatty acid, inhibited histone acetylation in DRGs and abolished K+ channel dysregulation-induced neuronal hyperexcitability after oxaliplatin treatment. Mechanical allodynia and cold hyperalgesia of mice receiving an accumulation of 15 mg/kg oxaliplatin, with or without intraperitoneal sodium butyrate supplementation, were assessed using von Frey test and acetone evaporation test. Differential expressions of histone deacetylases (HDACs) and pain-related K+ channels were quantified with rt-qPCR and protein assays. Immunofluorescence assays of histone acetylation at H3K9/14 were performed in primary DRG cultures treated with sodium butyrate. Current clamp recording of action potentials and persistent outward current of Twik-related-spinal cord K+ (TRESK) channel were recorded in DRG neurons with small diameters extract. Accompanied by mechanical allodynia and cold hyperalgesia, HDAC1 was upregulated in mice receiving oxaliplatin treatment. Sodium butyrate enhanced global histone acetylation at H3K9/14 in DRG neurons. In vivo sodium butyrate supplementation restored oxaliplatin-induced Kcnj9 and Kcnk18 expression and pain-related behaviors in mice for at least 14 days. Oxaliplatin-induced increase in action potentials frequencies and decrease in magnitudes of KCNK18-related current were reversed in mice receiving sodium butyrate supplementation. This study suggests that sodium butyrate was a useful agent to relieve oxaliplatin-mediated neuropathic pain.
Collapse
Affiliation(s)
- Idy H T Ho
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yidan Zou
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kele Luo
- The Chinese University of Hong Kong, Shenzhen, China
| | - Fenfen Qin
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yanjun Jiang
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Qian Li
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Tingting Jin
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xinyi Zhang
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Huarong Chen
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Likai Tan
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Lin Zhang
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Microbiota I Centre (MagIC), The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Tony Gin
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - William K K Wu
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; The Chinese University of Hong Kong, Shenzhen, China; State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Changyu Jiang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, China.
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
3
|
Cao M, Huang P, Xu LS, Zhang YH. Analysis of gut microbiota-derived metabolites regulating pituitary neuroendocrine tumors through network pharmacology. Front Pharmacol 2024; 15:1403864. [PMID: 39295931 PMCID: PMC11408289 DOI: 10.3389/fphar.2024.1403864] [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: 05/11/2024] [Accepted: 08/15/2024] [Indexed: 09/21/2024] Open
Abstract
Pituitary neuroendocrine tumors (PitNETs) are a special class of tumors of the central nervous system that are closely related to metabolism, endocrine functions, and immunity. In this study, network pharmacology was used to explore the metabolites and pharmacological mechanisms of PitNET regulation by gut microbiota. The metabolites of the gut microbiota were obtained from the gutMGene database, and the targets related to the metabolites and PitNETs were determined using public databases. A total of 208 metabolites were mined from the gutMGene database; 1,192 metabolite targets were screened from the similarity ensemble approach database; and 2,303 PitNET-related targets were screened from the GeneCards database. From these, 392 overlapping targets were screened between the metabolite and PitNET-related targets, and the intersection between these overlapping and gutMGene database targets (223 targets) were obtained as the core targets (43 targets). Using the protein-protein interaction (PPI) network analysis, Kyoto encyclopedia of genes and genomes (KEGG) signaling pathway and metabolic pathway analysis, CXCL8 was obtained as a hub target, tryptophan metabolism was found to be a key metabolic pathway, and IL-17 signaling was screened as the key KEGG signaling pathway. In addition, molecular docking analysis of the active metabolites and target were performed, and the results showed that baicalin, baicalein, and compound K had good binding activities with CXCL8. We also describe the potential mechanisms for treating PitNETs using the information on the microbiota (Bifidobacterium adolescentis), signaling pathway (IL-17), target (CXCL8), and metabolites (baicalin, baicalein, and compound K); we expect that these will provide a scientific basis for further study.
Collapse
Affiliation(s)
- Min Cao
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Ping Huang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Lun-Shan Xu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi-Hua Zhang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
4
|
Peng Y, Tun HM, Ng SC, Wai HKF, Zhang X, Parks J, Field CJ, Mandhane P, Moraes TJ, Simons E, Turvey SE, Subbarao P, Brook JR, Takaro TK, Scott JA, Chan FKL, Kozyrskyj AL. Maternal smoking during pregnancy increases the risk of gut microbiome-associated childhood overweight and obesity. Gut Microbes 2024; 16:2323234. [PMID: 38436093 PMCID: PMC10913716 DOI: 10.1080/19490976.2024.2323234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/21/2024] [Indexed: 03/05/2024] Open
Abstract
Childhood obesity is linked to maternal smoking during pregnancy. Gut microbiota may partially mediate this association and could be potential targets for intervention; however, its role is understudied. We included 1,592 infants from the Canadian Healthy Infants Longitudinal Development Cohort. Data on environmental exposure and lifestyle factors were collected prenatally and throughout the first three years. Weight outcomes were measured at one and three years of age. Stool samples collected at 3 and 12 months were analyzed by sequencing the V4 region of 16S rRNA to profile microbial compositions and magnetic resonance spectroscopy to quantify the metabolites. We showed that quitting smoking during pregnancy did not lower the risk of offspring being overweight. However, exclusive breastfeeding until the third month of age may alleviate these risks. We also reported that maternal smoking during pregnancy significantly increased Firmicutes abundance and diversity. We further revealed that Firmicutes diversity mediates the elevated risk of childhood overweight and obesity linked to maternal prenatal smoking. This effect possibly occurs through excessive microbial butyrate production. These findings add to the evidence that women should quit smoking before their pregnancies to prevent microbiome-mediated childhood overweight and obesity risk, and indicate the potential obesogenic role of excessive butyrate production in early life.
Collapse
Affiliation(s)
- Ye Peng
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Hein M Tun
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Siew C Ng
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Department of Medicine and Therapeutics, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Hogan Kok-Fung Wai
- HKU-Pasteur Research Pole, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Xi Zhang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Jaclyn Parks
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
- Cancer Control Research, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Catherine J Field
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Piush Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Theo J Moraes
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Elinor Simons
- Department of Pediatrics and Child Health, Children’s Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Stuart E Turvey
- Department of Pediatrics, Child and Family Research Institute, BC Children’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Padmaja Subbarao
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jeffrey R Brook
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Tim K Takaro
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Francis KL Chan
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
5
|
Anderson G. Melatonin, BAG-1 and cortisol circadian interactions in tumor pathogenesis and patterned immune responses. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:962-993. [PMID: 37970210 PMCID: PMC10645470 DOI: 10.37349/etat.2023.00176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/07/2023] [Indexed: 11/17/2023] Open
Abstract
A dysregulated circadian rhythm is significantly associated with cancer risk, as is aging. Both aging and circadian dysregulation show suppressed pineal melatonin, which is indicated in many studies to be linked to cancer risk and progression. Another independently investigated aspect of the circadian rhythm is the cortisol awakening response (CAR), which is linked to stress-associated hypothalamus-pituitary-adrenal (HPA) axis activation. CAR and HPA axis activity are primarily mediated via activation of the glucocorticoid receptor (GR), which drives patterned gene expression via binding to the promotors of glucocorticoid response element (GRE)-expressing genes. Recent data shows that the GR can be prevented from nuclear translocation by the B cell lymphoma-2 (Bcl-2)-associated athanogene 1 (BAG-1), which translocates the GR to mitochondria, where it can have diverse effects. Melatonin also suppresses GR nuclear translocation by maintaining the GR in a complex with heat shock protein 90 (Hsp90). Melatonin, directly and/or epigenetically, can upregulate BAG-1, suggesting that the dramatic 10-fold decrease in pineal melatonin from adolescence to the ninth decade of life will attenuate the capacity of night-time melatonin to modulate the effects of the early morning CAR. The interactions of pineal melatonin/BAG-1/Hsp90 with the CAR are proposed to underpin how aging and circadian dysregulation are associated with cancer risk. This may be mediated via differential effects of melatonin/BAG-1/Hsp90/GR in different cells of microenvironments across the body, from which tumors emerge. This provides a model of cancer pathogenesis that better integrates previously disparate bodies of data, including how immune cells are regulated by cancer cells in the tumor microenvironment, at least partly via the cancer cell regulation of the tryptophan-melatonin pathway. This has a number of future research and treatment implications.
Collapse
|
6
|
Karbownik MS, Sokołowska P, Kowalczyk E. Gut Microbiota Metabolites Differentially Release Gliotransmitters from the Cultured Human Astrocytes: A Preliminary Report. Int J Mol Sci 2023; 24:ijms24076617. [PMID: 37047602 PMCID: PMC10095279 DOI: 10.3390/ijms24076617] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Butyrate and indole-3-propionic acid represent the CNS-available gut microbiota metabolites exhibiting potentially beneficial effects on human brain function and being tested as antidepressants. Astrocytes represent one of the putative targets for the gut metabolites; however, the mechanism of action of butyrate and indole-3-propionic acid is not well understood. In order to test this mechanism, a human astrocyte cell-line culture was treated with the compounds or without them, and the supernatants were collected for the analysis of ATP and glutamate gliotransmitter release with the use of luminescent and fluorescent methods, respectively. A 10-min incubation of astrocytes with 1–5 mM butyrate increased the ATP gliotransmitter release by 78% (95%CI: 45–119%), p < 0.001. The effect was found to be mediated by the cytosolic Ca2+ mobilization. Both 10-min and 24-h treatments with indole-3-propionic acid produced no significant effects on the release of gliotransmitters. The results for glutamate release were inconclusive due to a specific glutamate release pattern discovered in the tested model. This preliminary report of butyrate-induced ATP gliotransmitter release appears to provide a novel mechanistic explanation for the beneficial effect of this gut microbiota metabolite on brain function; however, the results require further evaluation in more composed models.
Collapse
Affiliation(s)
- Michał Seweryn Karbownik
- Department of Pharmacology and Toxicology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland
| | - Paulina Sokołowska
- Department of Pharmacology and Toxicology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland
| |
Collapse
|
7
|
Taurine Stimulates AMP-Activated Protein Kinase and Modulates the Skeletal Muscle Functions in Rats via the Induction of Intracellular Calcium Influx. Int J Mol Sci 2023; 24:ijms24044125. [PMID: 36835534 PMCID: PMC9962205 DOI: 10.3390/ijms24044125] [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: 12/22/2022] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Taurine (2-aminoethanesulfonic acid) is a free amino acid abundantly found in mammalian tissues. Taurine plays a role in the maintenance of skeletal muscle functions and is associated with exercise capacity. However, the mechanism underlying taurine function in skeletal muscles has not yet been elucidated. In this study, to investigate the mechanism of taurine function in the skeletal muscles, the effects of short-term administration of a relatively low dose of taurine on the skeletal muscles of Sprague-Dawley rats and the underlying mechanism of taurine function in cultured L6 myotubes were investigated. The results obtained in this study in rats and L6 cells indicate that taurine modulates the skeletal muscle function by stimulating the expression of genes and proteins associated with mitochondrial and respiratory metabolism through the activation of AMP-activated protein kinase via the calcium signaling pathway.
Collapse
|
8
|
Zhang C, Xue P, Zhang H, Tan C, Zhao S, Li X, Sun L, Zheng H, Wang J, Zhang B, Lang W. Gut brain interaction theory reveals gut microbiota mediated neurogenesis and traditional Chinese medicine research strategies. Front Cell Infect Microbiol 2022; 12:1072341. [PMID: 36569198 PMCID: PMC9772886 DOI: 10.3389/fcimb.2022.1072341] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/07/2022] [Indexed: 12/13/2022] Open
Abstract
Adult neurogenesis is the process of differentiation of neural stem cells (NSCs) into neurons and glial cells in certain areas of the adult brain. Defects in neurogenesis can lead to neurodegenerative diseases, mental disorders, and other maladies. This process is directionally regulated by transcription factors, the Wnt and Notch pathway, the extracellular matrix, and various growth factors. External factors like stress, physical exercise, diet, medications, etc., affect neurogenesis and the gut microbiota. The gut microbiota may affect NSCs through vagal, immune and chemical pathways, and other pathways. Traditional Chinese medicine (TCM) has been proven to affect NSCs proliferation and differentiation and can regulate the abundance and metabolites produced by intestinal microorganisms. However, the underlying mechanisms by which these factors regulate neurogenesis through the gut microbiota are not fully understood. In this review, we describe the recent evidence on the role of the gut microbiota in neurogenesis. Moreover, we hypothesize on the characteristics of the microbiota-gut-brain axis based on bacterial phyla, including microbiota's metabolites, and neuronal and immune pathways while providing an outlook on TCM's potential effects on adult neurogenesis by regulating gut microbiota.
Collapse
Affiliation(s)
- Chenxi Zhang
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, China
| | - Peng Xue
- Medical School of Nantong University, Nantong University, Nantong, China
| | - Haiyan Zhang
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, China
| | - Chenxi Tan
- Department of Infection Control, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Shiyao Zhao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Xudong Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lihui Sun
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, China
| | - Huihui Zheng
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, China
| | - Jun Wang
- The Academic Affairs Office, Qiqihar Medical University, Qiqihar, China
| | - Baoling Zhang
- Department of Operating Room, Qiqihar First Hospital, Qiqihar, China
| | - Weiya Lang
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, China,*Correspondence: Weiya Lang,
| |
Collapse
|
9
|
Słupecka-Ziemilska M, Pierzynowski SG, Szczurek P, Pierzynowska K, Wychowański P, Seklecka B, Koperski M, Starzyńska A, Szkopek D, Donaldson J, Andrzejewski K, Woliński J. Milk Formula Enriched with Sodium Butyrate Influences Small Intestine Contractility in Neonatal Pigs. Nutrients 2022; 14:nu14204301. [PMID: 36296985 PMCID: PMC9608939 DOI: 10.3390/nu14204301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022] Open
Abstract
Butyrate, a by-product of gut bacteria fermentation as well as the digestion of fat in mother’s milk, exerts a wide spectrum of beneficial effects in the gastrointestinal tissues. The present study aimed to determine the effects of sodium butyrate on small intestine contractility in neonatal piglets. Piglets were fed milk formula alone (group C) or milk formula supplemented with sodium butyrate (group B). After a 7-day treatment period, isometric recordings of whole-thickness segments of the duodenum and middle jejunum were obtained by electric field stimulation under the influence of increasing doses of Ach (acetylocholine) in the presence of TTX (tetrodotoxin) and atropine. Moreover, structural properties of the intestinal wall were assessed, together with the expression of cholinergic and muscarinic receptors (M1 and M2). In both intestinal segments (duodenum and middle jejunum), EFS (electric field stimulation) impulses resulted in increased contractility and amplitude of contractions in group B compared to group C. Additionally, exposure to dietary butyrate led to a significant increase in tunica muscularis thickness in the duodenum, while mitotic and apoptotic indices were increased in the middle jejunum. The expression of M1 and M2 receptors in the middle jejunum was significantly higher after butyrate treatment. The results indicate increased cholinergic signaling and small intestinal growth and renewal in response to feeding with milk formula enriched with sodium butyrate in neonatal piglets.
Collapse
Affiliation(s)
- Monika Słupecka-Ziemilska
- Department of Human Epigenetics, Mossakowski Medical Research Institute Polish Academy of Sciences, 02-106 Warszawa, Poland
| | - Stefan Grzegorz Pierzynowski
- Department of Medical Biology, Institute of Rural Health, 20-090 Lublin, Poland
- SGP + Group, 231 32 Trelleborg, Sweden
- Department of Biology, Lund University, Sölvegatan 35, 223 62 Lund, Sweden
| | - Paulina Szczurek
- Department of Animal Nutrition and Feed Sciences, National Research Institute of Animal Production, 32-083 Balice, Poland
| | - Kateryna Pierzynowska
- Department of Biology, Lund University, Sölvegatan 35, 223 62 Lund, Sweden
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, 05-110 Jabłonna, Poland
- Correspondence: (K.P.); (J.W.)
| | - Piotr Wychowański
- Division of Oral Surgery and Implantology, Department of Head and Neck, Institute of Clinical Dentistry, Oral Surgery and Implantology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS-Universita Cattolica del Sacro Coure, 00168 Rome, Italy
| | | | - Maciej Koperski
- Department of Human Epigenetics, Mossakowski Medical Research Institute Polish Academy of Sciences, 02-106 Warszawa, Poland
| | - Anna Starzyńska
- Departament of Oral Surgery, Medical University of Gdańsk, 7 Dębinki Street, 80-211 Gdańsk, Poland
| | - Dominika Szkopek
- Large Animal Models Laboratory, The Kielanowski Institute of Animal Physiology and Nutrition, 05-110 Jabłonna, Poland
| | - Janine Donaldson
- SGP + Group, 231 32 Trelleborg, Sweden
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Krzysztof Andrzejewski
- Department of Orthopedics and Traumatology, Veteran’s Memorial Hospital, Medical University of Łódź, 90-549 Łódź, Poland
| | - Jarosław Woliński
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, 05-110 Jabłonna, Poland
- Large Animal Models Laboratory, The Kielanowski Institute of Animal Physiology and Nutrition, 05-110 Jabłonna, Poland
- Correspondence: (K.P.); (J.W.)
| |
Collapse
|
10
|
Rekha K, Venkidasamy B, Samynathan R, Nagella P, Rebezov M, Khayrullin M, Ponomarev E, Bouyahya A, Sarkar T, Shariati MA, Thiruvengadam M, Simal-Gandara J. Short-chain fatty acid: An updated review on signaling, metabolism, and therapeutic effects. Crit Rev Food Sci Nutr 2022; 64:2461-2489. [PMID: 36154353 DOI: 10.1080/10408398.2022.2124231] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Fatty acids are good energy sources (9 kcal per gram) that aerobic tissues can use except for the brain (glucose is an alternative source). Apart from the energy source, fatty acids are necessary for cell signaling, learning-related memory, modulating gene expression, and functioning as cytokine precursors. Short-chain fatty acids (SCFAs) are saturated fatty acids arranged as a straight chain consisting minimum of 6 carbon atoms. SCFAs possess various beneficial effects like improving metabolic function, inhibiting insulin resistance, and ameliorating immune dysfunction. In this review, we discussed the biogenesis, absorption, and transport of SCFA. SCFAs can act as signaling molecules by stimulating G protein-coupled receptors (GPCRs) and suppressing histone deacetylases (HDACs). The role of SCFA on glucose metabolism, fatty acid metabolism, and its effect on the immune system is also reviewed with updated details. SCFA possess anticancer, anti-diabetic, and hepatoprotective effects. Additionally, the association of protective effects of SCFA against brain-related diseases, kidney diseases, cardiovascular damage, and inflammatory bowel diseases were also reviewed. Nanotherapy is a branch of nanotechnology that employs nanoparticles at the nanoscale level to treat various ailments with enhanced drug stability, solubility, and minimal side effects. The SCFA functions as drug carriers, and nanoparticles were also discussed. Still, much research was not focused on this area. SCFA functions in host gene expression through inhibition of HDAC inhibition. However, the study has to be focused on the molecular mechanism of SCFA against various diseases that still need to be investigated.
Collapse
Affiliation(s)
- Kaliaperumal Rekha
- Department of Environmental and Herbal Science, Tamil University, Thanjavur, Tamil Nadu, India
| | - Baskar Venkidasamy
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | | | - Praveen Nagella
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, Karnataka, India
| | - Maksim Rebezov
- Department of Scientific Research, V. M. Gorbatov Federal Research Center for Food Systems, Moscow, Russia
- Department of Scientific Research, Russian State Agrarian University-Moscow Timiryazev Agricultural Academy, Moscow, Russia
- Department of Scientific Research, K. G. Razumovsky Moscow State University of technologies and management (The First Cossack University), Moscow, Russia
| | - Mars Khayrullin
- Department of Scientific Research, K. G. Razumovsky Moscow State University of technologies and management (The First Cossack University), Moscow, Russia
| | - Evgeny Ponomarev
- Department of Scientific Research, K. G. Razumovsky Moscow State University of technologies and management (The First Cossack University), Moscow, Russia
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Tanmay Sarkar
- Department of Food Processing Technology, Malda Polytechnic, West Bengal State Council of Technical Education, Government of West Bengal, Malda, West Bengal, India
| | - Mohammad Ali Shariati
- Department of Scientific Research, Russian State Agrarian University-Moscow Timiryazev Agricultural Academy, Moscow, Russia
- Department of Scientific Research, K. G. Razumovsky Moscow State University of technologies and management (The First Cossack University), Moscow, Russia
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Sciences, Konkuk University, Seoul, South Korea
| | - Jesus Simal-Gandara
- Analytical Chemistry and Food Science Department, Faculty of Science, Universidade de Vigo, Nutrition and Bromatology Group, Ourense, Spain
| |
Collapse
|
11
|
Lin K, Zhu L, Yang L. Gut and obesity/metabolic disease: Focus on microbiota metabolites. MedComm (Beijing) 2022; 3:e171. [PMID: 36092861 PMCID: PMC9437302 DOI: 10.1002/mco2.171] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/10/2022] Open
Abstract
Obesity is often associated with the risk of chronic inflammation and other metabolic diseases, such as diabetes, cardiovascular disease, and cancer. The composition and activity of the gut microbiota play an important role in this process, affecting a range of physiological processes, such as nutrient absorption and energy metabolism. The active gut microbiota can produce a large number of physiologically active substances during the process of intestinal metabolism and reproduction, including short-chain/long-chain fatty acids, secondary bile acids, and tryptophan metabolites with beneficial effects on metabolism, as well as negative metabolites, including trimethylamine N-oxide, delta-valerobetaine, and imidazole propionate. How gut microbiota specifically affect and participate in metabolic and immune activities, especially the metabolites directly produced by gut microbiota, has attracted extensive attention. So far, some animal and human studies have shown that gut microbiota metabolites are correlated with host obesity, energy metabolism, and inflammation. Some pathways and mechanisms are slowly being discovered. Here, we will focus on the important metabolites of gut microbiota (beneficial and negative), and review their roles and mechanisms in obesity and related metabolic diseases, hoping to provide a new perspective for the treatment and remission of obesity and other metabolic diseases from the perspective of metabolites.
Collapse
Affiliation(s)
- Ke Lin
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Lixin Zhu
- Guangdong Institute of GastroenterologyGuangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseaseSixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Colorectal SurgerySixth Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
12
|
Zamarbide M, Martinez-Pinilla E, Gil-Bea F, Yanagisawa M, Franco R, Perez-Mediavilla A. Genetic Inactivation of Free Fatty Acid Receptor 3 Impedes Behavioral Deficits and Pathological Hallmarks in the APP swe Alzheimer's Disease Mouse Model. Int J Mol Sci 2022; 23:ijms23073533. [PMID: 35408893 PMCID: PMC8999053 DOI: 10.3390/ijms23073533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 01/01/2023] Open
Abstract
The free fatty acid FFA3 receptor (FFA3R) belongs to the superfamily of G-protein-coupled receptors (GPCRs). In the intestine and adipose tissue, it is involved in the regulation of energy metabolism, but its function in the brain is unknown. We aimed, first, to investigate the expression of the receptor in the hippocampus of Alzheimer disease (AD) patients at different stages of the disease and, second, to assess whether genetic inactivation of the Ffar3 gene could affect the phenotypic features of the APPswe mouse model. The expression of transcripts for FFA receptors in postmortem human hippocampal samples and in the hippocampus of wild-type and transgenic mice was analyzed by RT-qPCR. We generated a double transgenic mouse, FFA3R−/−/APPswe, to perform cognition studies and to assess, by immunoblotting Aβ and tau pathologies and the differential expression of synaptic plasticity-related proteins. For the first time, the occurrence of the FFA3R in the human hippocampus and its overexpression, even in the first stages of AD, was demonstrated. Remarkably, FFA3R−/−/APPswe mice do not have the characteristic memory impairment of 12-month-old APPswe mice. Additionally, this newly generated transgenic line does not develop the most important Alzheimer’s disease (AD)-related features, such as amyloid beta (Aβ) brain accumulations and tau hyperphosphorylation. These findings are accompanied by increased levels of the insulin-degrading enzyme (IDE) and lower activity of the tau kinases GSK3β and Cdk5. We conclude that the brain FFA3R is involved in cognitive processes and that its inactivation prevents AD-like cognitive decline and pathological hallmarks.
Collapse
Affiliation(s)
- Marta Zamarbide
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (M.Z.); (E.M.-P.); (F.G.-B.)
- Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain
| | - Eva Martinez-Pinilla
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (M.Z.); (E.M.-P.); (F.G.-B.)
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33003 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Francisco Gil-Bea
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (M.Z.); (E.M.-P.); (F.G.-B.)
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba 305-8577, Japan;
| | - Rafael Franco
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (M.Z.); (E.M.-P.); (F.G.-B.)
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Network Center, Neurodegenerative Diseases, CiberNed, Spanish National Health Institute “Carlos III”, 28031 Madrid, Spain
- Correspondence: (R.F.); (A.P.-M.); Tel.: +34-934021208 (R.F.); +34-948194700 (ext. 2033) (A.P.-M.)
| | - Alberto Perez-Mediavilla
- Neuroscience Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain; (M.Z.); (E.M.-P.); (F.G.-B.)
- Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain
- Correspondence: (R.F.); (A.P.-M.); Tel.: +34-934021208 (R.F.); +34-948194700 (ext. 2033) (A.P.-M.)
| |
Collapse
|
13
|
Czernik PJ, Golonka RM, Chakraborty S, Yeoh BS, Abokor AA, Saha P, Yeo JY, Mell B, Cheng X, Baroi S, Tian Y, Patterson AD, Joe B, Vijay-Kumar M, Lecka-Czernik B. Reconstitution of the host holobiont in germ-free born male rats acutely increases bone growth and affects marrow cellular content. Physiol Genomics 2021; 53:518-533. [PMID: 34714176 PMCID: PMC8714805 DOI: 10.1152/physiolgenomics.00017.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 11/22/2022] Open
Abstract
Integration of microbiota in a host begins at birth and progresses during adolescence, forming a multidirectional system of physiological interactions. Here, we present an instantaneous effect of natural, bacterial gut colonization on the acceleration of longitudinal and radial bone growth in germ-free born, 7-wk-old male rats. Changes in bone mass and structure were analyzed after 10 days following the onset of colonization through cohousing with conventional rats and revealed unprecedented acceleration of bone accrual in cortical and trabecular compartments, increased bone tissue mineral density, improved proliferation and hypertrophy of growth plate chondrocytes, bone lengthening, and preferential deposition of periosteal bone in the tibia diaphysis. In addition, the number of small in size adipocytes increased, whereas the number of megakaryocytes decreased, in the bone marrow of conventionalized germ-free rats indicating that not only bone mass but also bone marrow environment is under control of gut microbiota signaling. The changes in bone status paralleled with a positive shift in microbiota composition toward short-chain fatty acids (SCFA)-producing microbes and a considerable increase in cecal SCFA concentrations, specifically butyrate. Furthermore, reconstitution of the host holobiont increased hepatic expression of IGF-1 and its circulating levels. Elevated serum levels of 25-hydroxy vitamin D and alkaline phosphatase pointed toward an active process of bone formation. The acute stimulatory effect on bone growth occurred independently of body mass increase. Overall, the presented model of conventionalized germ-free rats could be used to study microbiota-based therapeutics for combatting dysbiosis-related bone disorders.
Collapse
Affiliation(s)
- Piotr J Czernik
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Rachel M Golonka
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Saroj Chakraborty
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Beng San Yeoh
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Ahmed A Abokor
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Piu Saha
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Ji-Youn Yeo
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Blair Mell
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Xi Cheng
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Sudipta Baroi
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Yuan Tian
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Bina Joe
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Matam Vijay-Kumar
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Beata Lecka-Czernik
- Microbiome Consortium, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| |
Collapse
|
14
|
Liu S, Ma JY, Zhou J, Wu JD, Li JH, Alugongo GM, Xiao JX, Wang JJ, Wang YJ, Wang W, Li SL, Cao ZJ. Tributyrin supplementation in pasteurized waste milk: Effects on growth performance, health, and blood parameters of dairy calves. J Dairy Sci 2021; 104:12496-12507. [PMID: 34593232 DOI: 10.3168/jds.2021-20645] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/13/2021] [Indexed: 01/01/2023]
Abstract
This study evaluated the effects of incremental tributyrin supplementation in pasteurized waste milk on growth performance, health, and blood metabolism of dairy calves before and after weaning. Forty-eight newborn female Holstein dairy calves (39.6 ± 2.75 kg; mean ± standard deviation) were blocked by age and randomly assigned to 3 treatments: pasteurized waste milk (1) without supplementation, (2) with 1 g/L of tributyrin products (unprotected solid powder; containing 35% tributyrin), or (3) with 2 g/L of tributyrin products. The calves were weaned on d 56 and were raised until d 77. Data were analyzed for the preweaning, postweaning, and overall periods. The results showed that starter intake and hay intake were not different among treatments in any period of the trial, but the crude protein intake tended to increase linearly with tributyrin supplementation during the overall period. Although tributyrin supplementation had no effects on body weight during preweaning and overall periods, body weight increased linearly with tributyrin supplementation postweaning. The average daily gain tended to increase linearly during postweaning and overall periods. No effects were observed on feed efficiency in any period. A positive linear relationship between body length and tributyrin supplementation was observed during the postweaning period, but no differences were found for the other body structural measurements in any period. The results of diarrhea showed that tributyrin concentration had a negative linear relationship with diarrhea frequency during preweaning and overall periods. The rectal temperature did not differ among treatments in any period, but a treatment × week effect for rectal body temperature was observed. For blood metabolism, tributyrin supplementation had no effects on insulin, growth hormone, total protein, albumin, or globulin. No differences were found in serum amyloid A concentration in any of the periods, yet haptoglobin concentration decreased linearly with increasing tributyrin concentration during postweaning and overall periods. Endothelin concentration showed a tendency to decrease linearly during preweaning and postweaning periods and decreased linearly with tributyrin supplementation during the overall period. An increasing tributyrin concentration was associated with a negative linear relationship with IL-1β concentration during the preweaning period, and no differences were found in the other periods. The concentration of IL-6 and tumor necrosis factor α were not different among treatments in any of the periods. These data suggest that increasing the concentration of tributyrin in pasteurized waste milk could increase growth performance and health of dairy calves, and incremental tributyrin supplementation could linearly reduce haptoglobin, endothelin, and IL-1β concentrations, indicating a positive effect of tributyrin on alleviating oxidative stress and inflammatory status of dairy calves. Calves fed pasteurized waste milk supplemented with tributyrin products (containing 35% tributyrin) at 2 g/L compared with 1 g/L of milk had more improved growth and health.
Collapse
Affiliation(s)
- S Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - J Y Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - J Zhou
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - J D Wu
- Institute of Husbandry and Veterinary, Guizhou Provincial Academy of Agricultural Sciences, Guizhou 550005, P. R. China
| | - J H Li
- Department of Animal Science, University of California, Davis 95616
| | - G M Alugongo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - J X Xiao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - J J Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Y J Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - W Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - S L Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Z J Cao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China.
| |
Collapse
|
15
|
Chidambaram SB, Essa MM, Rathipriya AG, Bishir M, Ray B, Mahalakshmi AM, Tousif AH, Sakharkar MK, Kashyap RS, Friedland RP, Monaghan TM. Gut dysbiosis, defective autophagy and altered immune responses in neurodegenerative diseases: Tales of a vicious cycle. Pharmacol Ther 2021; 231:107988. [PMID: 34536490 DOI: 10.1016/j.pharmthera.2021.107988] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/16/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023]
Abstract
The human microbiota comprises trillions of symbiotic microorganisms and is involved in regulating gastrointestinal (GI), immune, nervous system and metabolic homeostasis. Recent observations suggest a bidirectional communication between the gut microbiota and the brain via immune, circulatory and neural pathways, termed the Gut-Brain Axis (GBA). Alterations in gut microbiota composition, such as seen with an increased number of pathobionts and a decreased number of symbionts, termed gut dysbiosis or microbial intestinal dysbiosis, plays a prominent role in the pathogenesis of central nervous system (CNS)-related disorders. Clinical reports confirm that GI symptoms often precede neurological symptoms several years before the development of neurodegenerative diseases (NDDs). Pathologically, gut dysbiosis disrupts the integrity of the intestinal barrier leading to ingress of pathobionts and toxic metabolites into the systemic circulation causing GBA dysregulation. Subsequently, chronic neuroinflammation via dysregulated immune activation triggers the accumulation of neurotoxic misfolded proteins in and around CNS cells resulting in neuronal death. Emerging evidence links gut dysbiosis to the aggravation and/or spread of proteinopathies from the peripheral nervous system to the CNS and defective autophagy-mediated proteinopathies. This review summarizes the current understanding of the role of gut microbiota in NDDs, and highlights a vicious cycle of gut dysbiosis, immune-mediated chronic neuroinflammation, impaired autophagy and proteinopathies, which contributes to the development of neurodegeneration in Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, amyotrophic lateral sclerosis and frontotemporal lobar degeneration. We also discuss novel therapeutic strategies targeting the modulation of gut dysbiosis through prebiotics, probiotics, synbiotics or dietary interventions, and faecal microbial transplantation (FMT) in the management of NDDs.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India.
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat 123, Oman; Ageing and Dementia Research Group, Sultan Qaboos University, Muscat 123, Oman; Biomedical Sciences Department, University of Pacific, Sacramento, CA, USA.
| | - A G Rathipriya
- Food and Brain Research Foundation, Chennai 600 094, Tamil Nadu, India
| | - Muhammed Bishir
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - A H Tousif
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Meena K Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - Rajpal Singh Kashyap
- Research Centre, Dr G. M. Taori Central India Institute of Medical Sciences (CIIMS), Nagpur, Maharashtra, India
| | - Robert P Friedland
- Department of Neurology, University of Louisville, Louisville, KY 40292, USA
| | - Tanya M Monaghan
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK; Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
16
|
García-Rodríguez D, Giménez-Cassina A. Ketone Bodies in the Brain Beyond Fuel Metabolism: From Excitability to Gene Expression and Cell Signaling. Front Mol Neurosci 2021; 14:732120. [PMID: 34512261 PMCID: PMC8429829 DOI: 10.3389/fnmol.2021.732120] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Ketone bodies are metabolites that replace glucose as the main fuel of the brain in situations of glucose scarcity, including prolonged fasting, extenuating exercise, or pathological conditions such as diabetes. Beyond their role as an alternative fuel for the brain, the impact of ketone bodies on neuronal physiology has been highlighted by the use of the so-called “ketogenic diets,” which were proposed about a century ago to treat infantile seizures. These diets mimic fasting by reducing drastically the intake of carbohydrates and proteins and replacing them with fat, thus promoting ketogenesis. The fact that ketogenic diets have such a profound effect on epileptic seizures points to complex biological effects of ketone bodies in addition to their role as a source of ATP. In this review, we specifically focus on the ability of ketone bodies to regulate neuronal excitability and their effects on gene expression to respond to oxidative stress. Finally, we also discuss their capacity as signaling molecules in brain cells.
Collapse
Affiliation(s)
- Darío García-Rodríguez
- Department of Molecular Biology, Centro de Biología Molecular "Severo Ochoa" (CBMSO UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Alfredo Giménez-Cassina
- Department of Molecular Biology, Centro de Biología Molecular "Severo Ochoa" (CBMSO UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
17
|
Zhao J, Hu J, Ma X. Sodium Decanoate Improves Intestinal Epithelial Barrier and Antioxidation via Activating G Protein-Coupled Receptor-43. Nutrients 2021; 13:nu13082756. [PMID: 34444916 PMCID: PMC8401944 DOI: 10.3390/nu13082756] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 01/01/2023] Open
Abstract
The study was conducted to explore actions of decanoic acid on regulating intestinal barrier and antioxidant functions in intestinal epithelium cells isolated from porcine jejunum (IPEC-J2) and C57/BL6 mice models. In vitro and vivo assays, mice and IPEC-J2 cells treated by H2O2 were disposed of sodium decanoate and sodium butyrate to determine intestinal barrier and antioxidant functions of the host. Results showed that sodium decanoate upregulated expression of tight junction proteins and improved antioxidant capacity in both IPEC-J2 cells treated by H2O2 and mice models (p < 0.05). Sodium decanoate increased weight gain and ileal villus height of mice compared with control and sodium butyrate treatments (p < 0.05). Sodium decanoate increased α-diversity of ileal microbiota, volatile fatty acids concentration, and G protein-coupled receptor-43 (GPR-43) expression in the ileum and colon of mice (p < 0.05). In conclusion, sodium decanoate improved antioxidant capacity, intestinal morphology, and gut physical barrier of intestinal epithelial cells, resulting in an increase growth performance of mice, which is mediated through activating GPR-43 signaling.
Collapse
Affiliation(s)
| | | | - Xi Ma
- Correspondence: ; Tel.: +86-10-62733588
| |
Collapse
|
18
|
Control of Protein and Energy Metabolism in the Pituitary Gland in Response to Three-Week Running Training in Adult Male Mice. Cells 2021; 10:cells10040736. [PMID: 33810540 PMCID: PMC8065971 DOI: 10.3390/cells10040736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/16/2021] [Accepted: 03/20/2021] [Indexed: 12/12/2022] Open
Abstract
It is assumed that crosstalk of central and peripheral tissues plays a role in the adaptive response to physical activity and exercise. Here, we wanted to study the effects of training and genetic predisposition in a marathon mouse model on mRNA expression in the pituitary gland. Therefore, we used a mouse model developed by phenotype selection for superior running performance (DUhTP) and non-inbred control mice (DUC). Both mouse lines underwent treadmill training for three weeks or were kept in a sedentary condition. In all groups, total RNA was isolated from the pituitary gland and sequenced. Molecular pathway analysis was performed by ingenuity pathway analysis (IPA). Training induced differential expression of 637 genes (DEGs) in DUC but only 50 DEGs in DUhTP mice. Genetic selection for enhanced running performance strongly affected gene expression in the pituitary gland and identified 1732 DEGs in sedentary DUC versus DUhTP mice. Training appeared to have an even stronger effect on gene expression in both lines and comparatively revealed 3828 DEGs in the pituitary gland. From the list of DEGs in all experimental groups, candidate genes were extracted by comparison with published genomic regions with significant effects on training responses in mice. Bioinformatic modeling revealed induction and coordinated expression of the pathways for ribosome synthesis and oxidative phosphorylation in DUC mice. By contrast, DUhTP mice were resistant to the positive effects of three-week training on protein and energy metabolism in the pituitary gland.
Collapse
|
19
|
Yu SY, Xu L. The interplay between host cellular and gut microbial metabolism in NAFLD development and prevention. J Appl Microbiol 2021; 131:564-582. [PMID: 33411984 DOI: 10.1111/jam.14992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/27/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Metabolism regulation centred on insulin resistance is increasingly important in nonalcoholic fatty liver disease (NAFLD). This review focuses on the interactions between the host cellular and gut microbial metabolism during the development of NAFLD. The cellular metabolism of essential nutrients, such as glucose, lipids and amino acids, is reconstructed with inflammation, immune mechanisms and oxidative stress, and these alterations modify the intestinal, hepatic and systemic environments, and regulate the composition and activity of gut microbes. Microbial metabolites, such as short-chain fatty acids, secondary bile acids, protein fermentation products, choline and ethanol and bacterial toxicants, such as lipopolysaccharides, peptidoglycans and bacterial DNA, play vital roles in NAFLD. The microbe-metabolite relationship is crucial for the modulation of intestinal microbial composition and metabolic activity. The intestinal microbiota and their metabolites participate in epithelial cell metabolism via a series of cell receptors and signalling pathways and remodel the metabolism of various cells in the liver via the gut-liver axis. Microbial metabolic manipulation is a promising strategy for NAFLD prevention, but larger-sampled clinical trials are required for future application.
Collapse
Affiliation(s)
- S-Y Yu
- Department of Gastroenterology, Ningbo First Hospital, Ningbo, China
| | - L Xu
- Department of Gastroenterology, Ningbo First Hospital, Ningbo, China
| |
Collapse
|
20
|
Maruta H, Yamashita H. Acetic acid stimulates G-protein-coupled receptor GPR43 and induces intracellular calcium influx in L6 myotube cells. PLoS One 2020; 15:e0239428. [PMID: 32997697 PMCID: PMC7526932 DOI: 10.1371/journal.pone.0239428] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022] Open
Abstract
Short chain fatty acids (SCFAs) produced endogenously in the gut by bacterial fermentation of dietary fiber have been studied as nutrients that act as signaling molecules to activate G-protein coupled receptors (GPCRs) such as GPR41 and GPR43. GPR43 functioning involves the suppression of lipid accumulation and maintaining body energy homeostasis, and is activated by acetic acid or propionic acid. Previously, we reported that the orally administered acetic acid improves lipid metabolism in liver and skeletal muscles and suppresses obesity, thus improving glucose tolerance. Acetic acid stimulates AMP-activated protein kinase (AMPK) through its metabolic pathway in skeletal muscle cells. We hypothesized that acetic acid would stimulate GPR43 in skeletal muscle cells and has function in modulating gene expression related to muscle characteristics through its signal pathway. The objective of the current study was to clarify this effect of acetic acid. The GPR43 expression, observed in the differentiated myotube cells, was increased upon acetic acid treatment. Acetic acid induced the intracellular calcium influx in the cells and this induction was significantly inhibited by the GPR43-specific siRNA treatment. The calcineurin molecule is activated by calcium/calmodulin and is associated with proliferation of slow-twitch fibers. Calcineurin was activated by acetic acid treatment and inhibited by the concomitant treatment with GPR43-siRNA. Acetic acid induced nuclear localization of myocyte enhancer factor 2A (MEF2A), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), and nuclear factor of activated t cells c1 (NFATc1). However, these localizations were abolished by the treatment with GPR43-siRNA. It was concluded that acetic acid plays a role in the activation of GPR43 and involves the proliferation of slow-twitch fibers in L6 skeletal muscles through the calcium-signaling pathway caused by induction of intracellular calcium influx.
Collapse
Affiliation(s)
- Hitomi Maruta
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, Japan
| | - Hiromi Yamashita
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, Japan
- Graduate School of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, Japan
- * E-mail:
| |
Collapse
|
21
|
Mizuta K, Sasaki H, Zhang Y, Matoba A, Emala CW. The short-chain free fatty acid receptor FFAR3 is expressed and potentiates contraction in human airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1248-L1260. [PMID: 32209026 DOI: 10.1152/ajplung.00357.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Emerging evidence suggests that gut microbiota-derived short-chain fatty acids (SCFAs; acetate, propionate, and butyrate) are important modulators of the inflammatory state in diseases such as asthma. However, the functional expression of the Gi protein-coupled free fatty acid receptors (FFAR2/GPR43 and FFAR3/GPR41) has not been identified on airway smooth muscle (ASM). Classically, acute activation of Gi-coupled receptors inhibits cyclic AMP (cAMP) synthesis, which impairs ASM relaxation and can also induce crosstalk between Gi- and Gq-signaling pathways, potentiating increases in intracellular Ca2+ concentration ([Ca2+]i), favoring ASM contraction. In contrast, chronic activation of Gi-coupled receptors can sensitize adenylyl cyclase resulting in increased cAMP synthesis favoring relaxation. We questioned whether the Gi-coupled FFAR2 or FFAR3 is expressed in human ASM, whether they modulate cAMP and [Ca2+]i, and whether SCFAs modulate human ASM tone. We detected the protein expression of FFAR3 but not FFAR2 in native human ASM and primary cultured human airway smooth muscle (HASM) cells. In HASM cells, acute activation of FFAR3 with SCFAs inhibited forskolin-stimulated cAMP accumulation, but chronic activation did not sensitize cAMP synthesis. SCFAs induced [Ca2+]i increases that were attenuated by pertussis toxin, gallein, U73122, or xestospongin C. Acute treatment with SCFAs potentiated acetylcholine-stimulated [Ca2+]i increases and stress fiber formation in cells and contraction of ex vivo human airway tissues. In contrast, chronic pretreatment of human ASM with propionate did not potentiate airway relaxation. Together, these findings demonstrate that FFAR3 is expressed in human ASM and contributes to ASM contraction via reduced cAMP and increased [Ca2+]i.
Collapse
Affiliation(s)
- Kentaro Mizuta
- Division of Dento-oral Anesthesiology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Haruka Sasaki
- Division of Dento-oral Anesthesiology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Yi Zhang
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Atsuko Matoba
- Division of Dento-oral Anesthesiology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Charles W Emala
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, New York
| |
Collapse
|
22
|
Silva YP, Bernardi A, Frozza RL. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front Endocrinol (Lausanne) 2020; 11:25. [PMID: 32082260 PMCID: PMC7005631 DOI: 10.3389/fendo.2020.00025] [Citation(s) in RCA: 1475] [Impact Index Per Article: 295.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
A substantial body of evidence supports that the gut microbiota plays a pivotal role in the regulation of metabolic, endocrine and immune functions. In recent years, there has been growing recognition of the involvement of the gut microbiota in the modulation of multiple neurochemical pathways through the highly interconnected gut-brain axis. Although amazing scientific breakthroughs over the last few years have expanded our knowledge on the communication between microbes and their hosts, the underpinnings of microbiota-gut-brain crosstalk remain to be determined. Short-chain fatty acids (SCFAs), the main metabolites produced in the colon by bacterial fermentation of dietary fibers and resistant starch, are speculated to play a key role in neuro-immunoendocrine regulation. However, the underlying mechanisms through which SCFAs might influence brain physiology and behavior have not been fully elucidated. In this review, we outline the current knowledge about the involvement of SCFAs in microbiota-gut-brain interactions. We also highlight how the development of future treatments for central nervous system (CNS) disorders can take advantage of the intimate and mutual interactions of the gut microbiota with the brain by exploring the role of SCFAs in the regulation of neuro-immunoendocrine function.
Collapse
Affiliation(s)
- Ygor Parladore Silva
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Andressa Bernardi
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Rudimar Luiz Frozza
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- *Correspondence: Rudimar Luiz Frozza
| |
Collapse
|
23
|
Wang H, Ren E, Xiang X, Su Y, Zhu W. Dynamic Changes in Serum Metabolomic Profiles of Growing Pigs Induced by Intravenous Infusion of Sodium Butyrate. Metabolites 2020; 10:metabo10010020. [PMID: 31906303 PMCID: PMC7023161 DOI: 10.3390/metabo10010020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/28/2019] [Accepted: 12/29/2019] [Indexed: 12/13/2022] Open
Abstract
This study aimed to explore the dynamic changes in metabolite profiles and metabolism pathways in the serum of growing pigs by intravenous infusion of sodium butyrate (SB). Fourteen crossbred growing barrows (BW = 23.70 ± 1.29 kg) fitted with jugular cannula were randomly allocated to the SB and control (Con) groups, each group consisted of seven replicates (pens), with one pig per pen. At 9:00 of each day during the experimental period, pigs in the SB group were infused with 10 mL of SB (200 mmol/L, pH 7.4, 37 °C) via precaval vein, while the Con group was treated with the same volume of physiological saline. On day 4, the blood of each pig was collected at 0, 30, 60, and 120 min after the intravenous infusion. Metabolites in the serum were detected by gas chromatograph-mass spectrometry analysis. Pathway analysis of metabolomic profiles showed that the differential metabolites mainly enriched in amino acid metabolism, lipid-related metabolism, and the tricarboxylic acid (TCA) cycle. More importantly, the relative concentrations of all eight essential amino acids, five non-essential amino acids, and two amino acid derivatives were decreased by the parenteral SB. In addition, SB significantly increased the relative concentrations of eicosanoic acid and octadecanoic acid and decreased the relative concentration of glycerol-3-phosphate at 0 min (three days after intravenous infusion of SB), which suggests that parenteral SB may increase stearates mobilization and decrease the biosynthesis of stearates. In conclusion, intravenous infusion of SB may induce more amino acids to synthesize proteins and affect fat metabolism through increasing fat mobilization and decreasing the biosynthesis of stearates. However, a further study is needed to understand the mechanism of extensive metabolic pathway changes induced by parenteral SB.
Collapse
Affiliation(s)
- Hongyu Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (H.W.); (E.R.); (W.Z.)
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Erdu Ren
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (H.W.); (E.R.); (W.Z.)
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoe Xiang
- National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, China;
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (H.W.); (E.R.); (W.Z.)
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
- Correspondence: ; Tel.: +86-25-84395523
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (H.W.); (E.R.); (W.Z.)
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
24
|
Wishart DS. Metabolomics for Investigating Physiological and Pathophysiological Processes. Physiol Rev 2019; 99:1819-1875. [PMID: 31434538 DOI: 10.1152/physrev.00035.2018] [Citation(s) in RCA: 598] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Metabolomics uses advanced analytical chemistry techniques to enable the high-throughput characterization of metabolites from cells, organs, tissues, or biofluids. The rapid growth in metabolomics is leading to a renewed interest in metabolism and the role that small molecule metabolites play in many biological processes. As a result, traditional views of metabolites as being simply the "bricks and mortar" of cells or just the fuel for cellular energetics are being upended. Indeed, metabolites appear to have much more varied and far more important roles as signaling molecules, immune modulators, endogenous toxins, and environmental sensors. This review explores how metabolomics is yielding important new insights into a number of important biological and physiological processes. In particular, a major focus is on illustrating how metabolomics and discoveries made through metabolomics are improving our understanding of both normal physiology and the pathophysiology of many diseases. These discoveries are yielding new insights into how metabolites influence organ function, immune function, nutrient sensing, and gut physiology. Collectively, this work is leading to a much more unified and system-wide perspective of biology wherein metabolites, proteins, and genes are understood to interact synergistically to modify the actions and functions of organelles, organs, and organisms.
Collapse
Affiliation(s)
- David S Wishart
- Departments of Biological Sciences and Computing Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
25
|
Weger BD, Rawashdeh O, Gachon F. At the Intersection of Microbiota and Circadian Clock: Are Sexual Dimorphism and Growth Hormones the Missing Link to Pathology? Bioessays 2019; 41:e1900059. [DOI: 10.1002/bies.201900059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/28/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Benjamin D. Weger
- Institute of Bioengineering, School of Life SciencesEcole Polytechnique Fédérale de LausanneLausanne CH‐1015 Switzerland
| | - Oliver Rawashdeh
- School of Biomedical Science, Faculty of MedicineThe University of QueenslandSt. Lucia QLD‐4072 Australia
| | - Frédéric Gachon
- Institute for Molecular BioscienceThe University of QueenslandSt. Lucia QLD‐4072 Australia
| |
Collapse
|
26
|
Lipid-regulating properties of butyric acid and 4-phenylbutyric acid: Molecular mechanisms and therapeutic applications. Pharmacol Res 2019; 144:116-131. [PMID: 30954630 DOI: 10.1016/j.phrs.2019.04.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/08/2019] [Accepted: 04/01/2019] [Indexed: 12/30/2022]
Abstract
In the past two decades, significant advances have been made in the etiology of lipid disorders. Concomitantly, the discovery of liporegulatory functions of certain short-chain fatty acids has generated interest in their clinical applications. In particular, butyric acid (BA) and its derivative, 4-phenylbutyric acid (PBA), which afford health benefits against lipid disorders while being generally well tolerated by animals and humans have been assessed clinically. This review examines the evidence from cell, animal and human studies pertaining to the lipid-regulating effects of BA and PBA, their molecular mechanisms and therapeutic potential. Collectively, the evidence supports the view that intakes of BA and PBA benefit lipid homeostasis across biological systems. We reviewed the evidence that BA and PBA downregulate de novo lipogenesis, ameliorate lipotoxicity, slow down atherosclerosis progression, and stimulate fatty acid β-oxidation. Central to their mode of action, BA appears to function as a histone deacetylase (HDAC) inhibitor while PBA acts as a chemical chaperone and/or a HDAC inhibitor. Areas of further inquiry include the effects of BA and PBA on adipogenesis, lipolysis and apolipoprotein metabolism.
Collapse
|
27
|
Chen DM, Zhang ML, Shi ZQ, Li CQ, Wang Q, Song JP, Xu Q, Li H, Zeng QP. Anti-inflammatory and Anti-infectious Dietary Paradigms May Be Crucial for Visceral Weight Reduction. Front Immunol 2019; 10:422. [PMID: 30906298 PMCID: PMC6418406 DOI: 10.3389/fimmu.2019.00422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 02/18/2019] [Indexed: 01/01/2023] Open
Affiliation(s)
- Dong-Mei Chen
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meng-Le Zhang
- School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhu-Qing Shi
- Science and Technology Industrial Park, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chang-Qing Li
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jian-Ping Song
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qin Xu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - He Li
- School of Basic Course, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing-Ping Zeng
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
28
|
Weger BD, Gobet C, Yeung J, Martin E, Jimenez S, Betrisey B, Foata F, Berger B, Balvay A, Foussier A, Charpagne A, Boizet-Bonhoure B, Chou CJ, Naef F, Gachon F. The Mouse Microbiome Is Required for Sex-Specific Diurnal Rhythms of Gene Expression and Metabolism. Cell Metab 2019; 29:362-382.e8. [PMID: 30344015 PMCID: PMC6370974 DOI: 10.1016/j.cmet.2018.09.023] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 06/27/2018] [Accepted: 09/25/2018] [Indexed: 02/08/2023]
Abstract
The circadian clock and associated feeding rhythms have a profound impact on metabolism and the gut microbiome. To what extent microbiota reciprocally affect daily rhythms of physiology in the host remains elusive. Here, we analyzed transcriptome and metabolome profiles of male and female germ-free mice. While mRNA expression of circadian clock genes revealed subtle changes in liver, intestine, and white adipose tissue, germ-free mice showed considerably altered expression of genes associated with rhythmic physiology. Strikingly, the absence of the microbiome attenuated liver sexual dimorphism and sex-specific rhythmicity. The resulting feminization of male and masculinization of female germ-free animals is likely caused by altered sexual development and growth hormone secretion, associated with differential activation of xenobiotic receptors. This defines a novel mechanism by which the microbiome regulates host metabolism.
Collapse
Affiliation(s)
- Benjamin D Weger
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Cédric Gobet
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland; Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jake Yeung
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Eva Martin
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Sonia Jimenez
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Bertrand Betrisey
- Cellular Metabolism, Department of Cell Biology, Nestlé Institute of Health Sciences, Nestlé Research, 1015 Lausanne, Switzerland
| | - Francis Foata
- Host-Microbe Interaction, Department of Gastro-Intestinal Health, Nestlé Institute of Health Sciences, Nestlé Research, 1000 Lausanne, Switzerland
| | - Bernard Berger
- Host-Microbe Interaction, Department of Gastro-Intestinal Health, Nestlé Institute of Health Sciences, Nestlé Research, 1000 Lausanne, Switzerland
| | - Aurélie Balvay
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Anne Foussier
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Aline Charpagne
- Genomics, Department of Multi-Omics, Nestlé Institute of Health Sciences, Nestlé Research, 1015 Lausanne, Switzerland
| | - Brigitte Boizet-Bonhoure
- Institut de Génétique Humaine, CNRS-Université de Montpellier UMR9002, 34396 Montpellier, France
| | - Chieh Jason Chou
- Host-Microbe Interaction, Department of Gastro-Intestinal Health, Nestlé Institute of Health Sciences, Nestlé Research, 1000 Lausanne, Switzerland
| | - Felix Naef
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Frédéric Gachon
- Department of Diabetes and Circadian Rhythms, Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
29
|
Frieten D, Gerbert C, Koch C, Dusel G, Eder K, Hoeflich A, Mielenz B, Hammon H. Influence of ad libitum milk replacer feeding and butyrate supplementation on the systemic and hepatic insulin-like growth factor I and its binding proteins in Holstein calves. J Dairy Sci 2018; 101:1661-1672. [DOI: 10.3168/jds.2017-13603] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/28/2017] [Indexed: 01/01/2023]
|
30
|
Liu H, Wang J, He T, Becker S, Zhang G, Li D, Ma X. Butyrate: A Double-Edged Sword for Health? Adv Nutr 2018; 9:21-29. [PMID: 29438462 PMCID: PMC6333934 DOI: 10.1093/advances/nmx009] [Citation(s) in RCA: 659] [Impact Index Per Article: 94.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 11/11/2017] [Indexed: 02/06/2023] Open
Abstract
Butyrate, a four-carbon short-chain fatty acid, is produced through microbial fermentation of dietary fibers in the lower intestinal tract. Endogenous butyrate production, delivery, and absorption by colonocytes have been well documented. Butyrate exerts its functions by acting as a histone deacetylase (HDAC) inhibitor or signaling through several G protein-coupled receptors (GPCRs). Recently, butyrate has received particular attention for its beneficial effects on intestinal homeostasis and energy metabolism. With anti-inflammatory properties, butyrate enhances intestinal barrier function and mucosal immunity. However, the role of butyrate in obesity remains controversial. Growing evidence has highlighted the impact of butyrate on the gut-brain axis. In this review, we summarize the present knowledge on the properties of butyrate, especially its potential effects and mechanisms involved in intestinal health and obesity.
Collapse
Affiliation(s)
- Hu Liu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ji Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ting He
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Sage Becker
- Department of Animal Science, Oklahoma State University, Stillwater, OK; Departments of Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, OK; Departments of Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Defa Li
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX,Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX,Address correspondence to XM (e-mail: )
| |
Collapse
|
31
|
Xie G, Zhou Q, Qiu CZ, Dai WK, Wang HP, Li YH, Liao JX, Lu XG, Lin SF, Ye JH, Ma ZY, Wang WJ. Ketogenic diet poses a significant effect on imbalanced gut microbiota in infants with refractory epilepsy. World J Gastroenterol 2017; 23:6164-6171. [PMID: 28970732 PMCID: PMC5597508 DOI: 10.3748/wjg.v23.i33.6164] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/09/2017] [Accepted: 07/12/2017] [Indexed: 02/07/2023] Open
Abstract
AIM To investigate whether patients with refractory epilepsy and healthy infants differ in gut microbiota (GM), and how ketogenic diet (KD) alters GM. METHODS A total of 14 epileptic and 30 healthy infants were recruited and seizure frequencies were recorded. Stool samples were collected for 16S rDNA sequencing using the Illumina Miseq platform. The composition of GM in each sample was analyzed with MOTHUR, and inter-group comparison was conducted by R software. RESULTS After being on KD treatment for a week, 64% of epileptic infants showed an obvious improvement, with a 50% decrease in seizure frequency. GM structure in epileptic infants (P1 group) differed dramatically from that in healthy infants (Health group). Proteobacteria, which had accumulated significantly in the P1 group, decreased dramatically after KD treatment (P2 group). Cronobacter predominated in the P1 group and remained at a low level both in the Health and P2 groups. Bacteroides increased significantly in the P2 group, in which Prevotella and Bifidobacterium also grew in numbers and kept increasing. CONCLUSION GM pattern in healthy infants differed dramatically from that of the epileptic group. KD could significantly modify symptoms of epilepsy and reshape the GM of epileptic infants.
Collapse
Affiliation(s)
- Gan Xie
- Department of Respiratory Medicine, Shenzhen Children’s Hospital, Shenzhen 518026, Guangdong Province, China
| | - Qian Zhou
- WeHealthGene Institute, Shenzhen 518129, Guangdong Province, China
| | - Chuang-Zhao Qiu
- WeHealthGene Institute, Shenzhen 518129, Guangdong Province, China
| | - Wen-Kui Dai
- WeHealthGene Institute, Shenzhen 518129, Guangdong Province, China
| | - He-Ping Wang
- Department of Respiratory Medicine, Shenzhen Children’s Hospital, Shenzhen 518026, Guangdong Province, China
| | - Yin-Hu Li
- WeHealthGene Institute, Shenzhen 518129, Guangdong Province, China
| | - Jian-Xiang Liao
- Department of Pediatric Neurology, Shenzhen Children’s Hospital, Shenzhen 518026, Guangdong Province, China
| | - Xin-Guo Lu
- Department of Pediatric Neurology, Shenzhen Children’s Hospital, Shenzhen 518026, Guangdong Province, China
| | - Su-Fang Lin
- Department of Pediatric Neurology, Shenzhen Children’s Hospital, Shenzhen 518026, Guangdong Province, China
| | - Jing-Hua Ye
- Department of Pediatric Neurology, Shenzhen Children’s Hospital, Shenzhen 518026, Guangdong Province, China
| | - Zhuo-Ya Ma
- Department of Respiratory Medicine, Shenzhen Children’s Hospital, Shenzhen 518026, Guangdong Province, China
| | - Wen-Jian Wang
- Department of Respiratory Medicine, Shenzhen Children’s Hospital, Shenzhen 518026, Guangdong Province, China
| |
Collapse
|
32
|
Butyrate influences intracellular levels of adenine and adenine derivatives in the fungus Penicillium restrictum. Microbiol Res 2017; 197:1-8. [PMID: 28219521 DOI: 10.1016/j.micres.2016.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/21/2016] [Accepted: 12/30/2016] [Indexed: 01/01/2023]
Abstract
Butyrate, a small fatty acid, has an important role in the colon of ruminants and mammalians including the inhibition of inflammation and the regulation of cell proliferation. There is also growing evidence that butyrate is influencing the histone structure in mammalian cells by inhibition of histone deacetylation. Butyrate shows furthermore an antimicrobial activity against fungi, yeast and bacteria, which is linked to its toxicity at a high concentration. In fungi there are indications that butyrate induces the production of secondary metabolites potentially via inhibition of histone deacetylases. However, information about the influence of butyrate on growth, primary metabolite production and metabolism, besides lipid catabolism, in fungi is scarce. We have identified the filamentous fungus Penicillium (P.) restrictum as a susceptible target for butyrate treatment in an antimicrobial activity screen. The antimicrobial activity was detected only in the mycelium of the butyrate treated culture. We investigated the effect of butyrate ranging from low (0.001mM) to high (30mM), potentially toxic, concentrations on biomass and antimicrobial activity. Butyrate at high concentrations (3 and 30mM) significantly reduced the fungal biomass. In contrast P. restrictum treated with 0.03mM of butyrate showed the highest antimicrobial activity. We isolated three antimicrobial active compounds, active against Staphylococcus aureus, from P. restrictum cellular extracts treated with butyrate: adenine, its derivate hypoxanthine and the nucleoside derivate adenosine. Production of all three compounds was increased at low butyrate concentrations. Furthermore we found that butyrate influences the intracellular level of the adenine nucleoside derivate cAMP, an important signalling molecule in fungi and various organisms. In conclusion butyrate treatment increases the intracellular levels of adenine and its respective derivatives.
Collapse
|
33
|
Mirković B, Murray MA, Lavelle GM, Molloy K, Azim AA, Gunaratnam C, Healy F, Slattery D, McNally P, Hatch J, Wolfgang M, Tunney MM, Muhlebach MS, Devery R, Greene CM, McElvaney NG. The Role of Short-Chain Fatty Acids, Produced by Anaerobic Bacteria, in the Cystic Fibrosis Airway. Am J Respir Crit Care Med 2015; 192:1314-24. [PMID: 26266556 PMCID: PMC4731701 DOI: 10.1164/rccm.201505-0943oc] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/12/2015] [Indexed: 12/22/2022] Open
Abstract
RATIONALE Anaerobic bacteria are present in large numbers in the airways of people with cystic fibrosis (PWCF). In the gut, anaerobes produce short-chain fatty acids (SCFAs) that modulate immune and inflammatory processes. OBJECTIVES To investigate the capacity of anaerobes to contribute to cystic fibrosis (CF) airway pathogenesis via SCFAs. METHODS Samples of 109 PWCF were processed using anaerobic microbiological culture with bacteria present identified by 16S RNA sequencing. SCFA levels in anaerobic supernatants and bronchoalveolar lavage (BAL) were determined by gas chromatography. The mRNA and/or protein expression of two SCFA receptors, GPR41 and GPR43, in CF and non-CF bronchial brushings and 16HBE14o(-) and CFBE41o(-) cells were evaluated using reverse transcription polymerase chain reaction, Western blot analysis, laser scanning cytometry, and confocal microscopy. SCFA-induced IL-8 secretion was monitored by ELISA. MEASUREMENTS AND MAIN RESULTS Fifty-seven (52.3%) of 109 PWCF were anaerobe positive. Prevalence increased with age, from 33.3% to 57.7% in PWCF younger (n = 24) and older (n = 85) than 6 years of age. All evaluated anaerobes produced millimolar concentrations of SCFAs, including acetic, propionic, and butyric acids. SCFA levels were higher in BAL samples of adults than in those of children. GPR41 levels were elevated in CFBE41o(-) versus 16HBE14o(-) cells; CF versus non-CF bronchial brushings; and 16HBE14o(-) cells after treatment with cystic fibrosis transmembrane conductance regulator inhibitor CFTR(inh)-172, CF BAL, or inducers of endoplasmic reticulum stress. SCFAs induced a dose-dependent and pertussis toxin-sensitive IL-8 response in bronchial epithelial cells, with a higher production of IL-8 in CFBE41o(-) than in 16HBE14o(-) cells. CONCLUSIONS This study illustrates that SCFAs contribute to excessive production of IL-8 in CF airways colonized with anaerobes via up-regulated GPR41.
Collapse
Affiliation(s)
- Bojana Mirković
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Michelle A. Murray
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Gillian M. Lavelle
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Kevin Molloy
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Ahmed Abdul Azim
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Cedric Gunaratnam
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Fiona Healy
- Temple Street Children’s University Hospital, Dublin, Ireland
| | | | - Paul McNally
- Our Lady’s Children’s Hospital, Crumlin, Dublin, Ireland
| | - Joe Hatch
- Cystic Fibrosis/Pulmonary Research and Treatment Center
- Department of Microbiology and Immunology, and
| | - Matthew Wolfgang
- Cystic Fibrosis/Pulmonary Research and Treatment Center
- Department of Microbiology and Immunology, and
| | - Michael M. Tunney
- CF & Airways Microbiology Group and
- School of Pharmacy, Queen’s University Belfast, Belfast, United Kingdom; and
| | - Marianne S. Muhlebach
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Rosaleen Devery
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Catherine M. Greene
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Noel G. McElvaney
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
34
|
Wang X, He G, Peng Y, Zhong W, Wang Y, Zhang B. Sodium butyrate alleviates adipocyte inflammation by inhibiting NLRP3 pathway. Sci Rep 2015; 5:12676. [PMID: 26234821 PMCID: PMC4522654 DOI: 10.1038/srep12676] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 07/06/2015] [Indexed: 02/06/2023] Open
Abstract
Insulin resistance (IR) is a common feature of Type II diabetes, metabolic disorders, hypertension and other vascular diseases. Recent studies showed that obesity-induced inflammation may be critical for IR. To investigate the anti-inflammatory effect of sodium butyrate (NaB) on obesity-induced inflammation, the db/db mice were intraperitoneally injected with NaB for 6 weeks. Glucose control was evaluated by glucose tolerance test (GTT) and insulin tolerance test (ITT). Adipose tissue was harvested for gene expression analysis. 3T3-L1 adipocytes were treated with Tnf-α to mimic the inflammatory state and gene expression was detected by realtime PCR and Western blotting. Our results showed that NaB treatment improved glucose control in db/db mice as determined by GTT and ITT tests. Gene expression analysis showed that NaB inhibited cytokines and immunological markers including CD68, Interferon-γ and Mcp in adipose tissues in db/db mice. Moreover, NaB inhibited cytokine releasing in 3T3-L1 adipocytes treated with TNF-α. Further analysis of inflammation pathway showed that NLRP3 was activated in db/db mice, which was efficiently inhibited by NaB treatment. Our data suggest that inhibition of obesity-induced inflammation alleviates IR, and NaB might be a potential anti-inflammatory agent for obesity.
Collapse
Affiliation(s)
- Xukai Wang
- Department of Cardiovascular Internal Medicine, Institute of Field Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Gang He
- Department of Medical Genetics, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Yan Peng
- Department of Cardiovascular Internal Medicine, Institute of Field Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Weitian Zhong
- Department of Cardiovascular Internal Medicine, Institute of Field Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan Wang
- Department of Medical Genetics, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Bo Zhang
- Department of Medical Genetics, College of Basic Medicine, Third Military Medical University, Chongqing, China
| |
Collapse
|
35
|
Wu S, Yi J, Zhang YG, Zhou J, Sun J. Leaky intestine and impaired microbiome in an amyotrophic lateral sclerosis mouse model. Physiol Rep 2015; 3:3/4/e12356. [PMID: 25847918 PMCID: PMC4425962 DOI: 10.14814/phy2.12356] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Emerging evidence has demonstrated that intestinal homeostasis and the microbiome play essential roles in neurological diseases, such as Parkinson's disease. Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by a progressive loss of motor neurons and muscle atrophy. Currently, there is no effective treatment. Most patients die within 3–5 years due to respiratory paralysis. Although the death of motor neurons is a hallmark of ALS, other organs may also contribute to the disease progression. We examined the gut of an ALS mouse model, G93A, which expresses mutant superoxide dismutase (SOD1G93A), and discovered a damaged tight junction structure and increased permeability with a significant reduction in the expression levels of tight junction protein ZO-1 and the adherens junction protein E-cadherin. Furthermore, our data demonstrated increased numbers of abnormal Paneth cells in the intestine of G93A mice. Paneth cells are specialized intestinal epithelial cells that can sense microbes and secrete antimicrobial peptides, thus playing key roles in host innate immune responses and shaping the gut microbiome. A decreased level of the antimicrobial peptides defensin 5 alpha was indeed found in the ALS intestine. These changes were associated with a shifted profile of the intestinal microbiome, including reduced levels of Butyrivibrio Fibrisolvens, Escherichia coli, and Fermicus, in G93A mice. The relative abundance of bacteria was shifted in G93A mice compared to wild-type mice. Principal coordinate analysis indicated a difference in fecal microbial communities between ALS and wild-type mice. Taken together, our study suggests a potential novel role of the intestinal epithelium and microbiome in the progression of ALS.
Collapse
Affiliation(s)
- Shaoping Wu
- Department of Biochemistry, Rush University Medical Center, Chicago, Illinois
| | - Jianxun Yi
- Department of Physiology, Kansas City University of Medicine and Bioscience, Kansas, Missouri
| | - Yong-Guo Zhang
- Department of Biochemistry, Rush University Medical Center, Chicago, Illinois
| | - Jingsong Zhou
- Department of Physiology, Kansas City University of Medicine and Bioscience, Kansas, Missouri
| | - Jun Sun
- Department of Biochemistry, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|