1
|
Huang Y, Wang YF, Miao J, Zheng RF, Li JY. Short-chain fatty acids: Important components of the gut-brain axis against AD. Biomed Pharmacother 2024; 175:116601. [PMID: 38749177 DOI: 10.1016/j.biopha.2024.116601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 06/03/2024] Open
Abstract
Alzheimer's disease (AD) comprises a group of neurodegenerative disorders with some changes in the brain, which could lead to the deposition of certain proteins and result in the degeneration and death of brain cells. Patients with AD manifest primarily as cognitive decline, psychiatric symptoms, and behavioural disorders. Short-chain fatty acids (SCFAs) are a class of saturated fatty acids (SFAs) produced by gut microorganisms through the fermentation of dietary fibre ingested. SCFAs, as a significant mediator of signalling, can have diverse physiological and pathological roles in the brain through the gut-brain axis, and play a positive effect on AD via multiple pathways. Firstly, differences in SCFAs and microbial changes have been stated in AD cases of humans and mice in this paper. And then, mechanisms of three main SCFAs in treating with AD have been summarized, as well as differences of gut bacteria. Finally, functions of SCFAs played in regulating intestinal flora homeostasis, modulating the immune system, and the metabolic system, which were considered to be beneficial for the treatment of AD, have been elucidated, and the key roles of gut bacteria and SCFAs were pointed out. All in all, this paper provides an overview of SCFAs and gut bacteria in AD, and can help people to understand the importance of gut-brain axis in AD.
Collapse
Affiliation(s)
- Yan Huang
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China
| | - Yi Feng Wang
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China
| | - Jing Miao
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China; Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830004, China.
| | - Rui Fang Zheng
- Xinjiang Key Laboratory of Uygur Medical Research, Xinjiang Institute of Materia Medica, Urumqi 830004, China.
| | - Jin Yao Li
- College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University, Urumqi 830004, China.
| |
Collapse
|
2
|
Karim MR, Iqbal S, Mohammad S, Morshed MN, Haque MA, Mathiyalagan R, Yang DC, Kim YJ, Song JH, Yang DU. Butyrate's (a short-chain fatty acid) microbial synthesis, absorption, and preventive roles against colorectal and lung cancer. Arch Microbiol 2024; 206:137. [PMID: 38436734 DOI: 10.1007/s00203-024-03834-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/28/2023] [Accepted: 01/04/2024] [Indexed: 03/05/2024]
Abstract
Butyrate, a short-chain fatty acid (SCFA) produced by bacterial fermentation of fiber in the colon, is a source of energy for colonocytes. Butyrate is essential for improving gastrointestinal (GI) health since it helps colonocyte function, reduces inflammation, preserves the gut barrier, and fosters a balanced microbiome. Human colonic butyrate producers are Gram-positive firmicutes, which are phylogenetically varied. The two most prevalent subgroups are associated with Eubacterium rectale/Roseburia spp. and Faecalibacterium prausnitzii. Now, the mechanism for the production of butyrate from microbes is a very vital topic to know. In the present study, we discuss the genes encoding the core of the butyrate synthesis pathway and also discuss the butyryl-CoA:acetate CoA-transferase, instead of butyrate kinase, which usually appears to be the enzyme that completes the process. Recently, butyrate-producing microbes have been genetically modified by researchers to increase butyrate synthesis from microbes. The activity of butyrate as a histone deacetylase inhibitor (HDACi) has led to several clinical trials to assess its effectiveness as a potential cancer treatment. Among various significant roles, butyrate is the main energy source for intestinal epithelial cells, which helps maintain colonic homeostasis. Moreover, people with non-small-cell lung cancer (NSCLC) have distinct gut microbiota from healthy adults and frequently have dysbiosis of the butyrate-producing bacteria in their guts. So, with an emphasis on colon and lung cancer, this review also discusses how the microbiome is crucial in preventing the progression of certain cancers through butyrate production. Further studies should be performed to investigate the underlying mechanisms of how these specific butyrate-producing bacteria can control both colon and lung cancer progression and prognosis.
Collapse
Affiliation(s)
- Md Rezaul Karim
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia, 7003, Bangladesh
| | - Safia Iqbal
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
- Department of Microbiology, Varendra Institute of Biosciences, Affiliated University of Rajshahi, Natore, 6400, Rajshahi, Bangladesh
| | - Shahnawaz Mohammad
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
| | - Md Niaj Morshed
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
| | - Md Anwarul Haque
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia, 7003, Bangladesh
| | - Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
| | - Deok Chun Yang
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
- Hanbangbio Inc., Yongin-Si, 17104, Gyeonggi-Do, Republic of Korea
| | - Yeon Ju Kim
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea
| | - Joong Hyun Song
- Department of Veterinary International Medicine, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea.
| | - Dong Uk Yang
- Department of Biopharmaceutical Biotechnology, College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Korea.
- AIBIOME, 6, Jeonmin-Ro 30Beon-Gil, Yuseong-Gu, Daejeon, Republic of Korea.
| |
Collapse
|
3
|
Del Negro I, Pez S, Versace S, Marziali A, Gigli GL, Tereshko Y, Valente M. Impact of Disease-Modifying Therapies on Gut-Brain Axis in Multiple Sclerosis. MEDICINA (KAUNAS, LITHUANIA) 2023; 60:6. [PMID: 38276041 PMCID: PMC10818907 DOI: 10.3390/medicina60010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024]
Abstract
Multiple sclerosis is a chronic, autoimmune-mediated, demyelinating disease whose pathogenesis remains to be defined. In past years, in consideration of a constantly growing number of patients diagnosed with multiple sclerosis, the impacts of different environmental factors in the pathogenesis of the disease have been largely studied. Alterations in gut microbiome composition and intestinal barrier permeability have been suggested to play an essential role in the regulation of autoimmunity. Thus, increased efforts are being conducted to demonstrate the complex interplay between gut homeostasis and disease pathogenesis. Numerous results confirm that disease-modifying therapies (DMTs) used for the treatment of MS, in addition to their immunomodulatory effect, could exert an impact on the intestinal microbiota, contributing to the modulation of the immune response itself. However, to date, the direct influence of these treatments on the microbiota is still unclear. This review intends to underline the impact of DMTs on the complex system of the microbiota-gut-brain axis in patients with multiple sclerosis.
Collapse
Affiliation(s)
- Ilaria Del Negro
- Clinical Neurology Unit, Udine University Hospital, Piazzale S. Maria della Misericordia, 33100 Udine, Italy
- Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy
| | - Sara Pez
- Clinical Neurology Unit, Udine University Hospital, Piazzale S. Maria della Misericordia, 33100 Udine, Italy
- Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy
| | - Salvatore Versace
- Clinical Neurology Unit, Udine University Hospital, Piazzale S. Maria della Misericordia, 33100 Udine, Italy
- Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy
| | - Alessandro Marziali
- Clinical Neurology Unit, Udine University Hospital, Piazzale S. Maria della Misericordia, 33100 Udine, Italy
- Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy
| | - Gian Luigi Gigli
- Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy
| | - Yan Tereshko
- Clinical Neurology Unit, Udine University Hospital, Piazzale S. Maria della Misericordia, 33100 Udine, Italy
- Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy
| | - Mariarosaria Valente
- Clinical Neurology Unit, Udine University Hospital, Piazzale S. Maria della Misericordia, 33100 Udine, Italy
- Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy
| |
Collapse
|
4
|
Zhang Z, Lu W, Liu P, Li M, Ge X, Yu B, Wu Z, Liu G, Ding N, Cui B, Chen X. Microbial modifications with Lycium barbarum L. oligosaccharides decrease hepatic fibrosis and mitochondrial abnormalities in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155068. [PMID: 37690228 DOI: 10.1016/j.phymed.2023.155068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/03/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Lycium barbarum L. is a typical Chinese herbal and edible plant and are now consumed globally. Low molecular weight L. barbarum L. oligosaccharides (LBO) exhibit better antioxidant activity and gastrointestinal digestibility in vitro than high molecular weight polysaccharides. However, the LBO on the treatment of liver disease is not studied. PURPOSE Modification of the gut microbial ecosystem by LBO is a promising treatment for liver fibrosis. STUDY DESIGN AND METHODS Herein, LBO were prepared and characterized. CCl4-treated mice were orally gavaged with LBO and the effects on hepatic fibrosis and mitochondrial abnormalities were evaluated according to relevant indicators (gut microbiota, faecal metabolites, and physiological and biochemical indices). RESULTS The results revealed that LBO, a potential prebiotic source, is a pyranose cyclic oligosaccharide possessing α-glycosidic and β-glycosidic bonds. Moreover, LBO supplementation restored the configuration of the bacterial community, enhanced the proliferation of beneficial species in the gastrointestinal tract (e.g., Bacillus, Tyzzerella, Fournierella and Coriobacteriaceae UCG-002), improved microbial metabolic alterations (i.e., carbohydrate metabolism, vitamin metabolism and entero-hepatic circulation), and increased antioxidants, including doxepin, in mice. Finally, LBO administration reduced serum inflammatory cytokine and hepatic hydroxyproline levels, improved intestinal and hepatic mitochondrial functions, and ameliorated mouse liver fibrosis. CONCLUSION These findings indicate that LBO can be utilized as a prebiotic and has a remarkable ability to mitigate liver fibrosis.
Collapse
Affiliation(s)
- Zheng Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Wenjia Lu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Pengfei Liu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Mengjie Li
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Xinyi Ge
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Bin Yu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Zhengzong Wu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Guimei Liu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Nannan Ding
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Bo Cui
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China.
| | - Xiao Chen
- College of Health Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250353, China.
| |
Collapse
|
5
|
Bresciani G, Manai F, Davinelli S, Tucci P, Saso L, Amadio M. Novel potential pharmacological applications of dimethyl fumarate-an overview and update. Front Pharmacol 2023; 14:1264842. [PMID: 37745068 PMCID: PMC10512734 DOI: 10.3389/fphar.2023.1264842] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Dimethyl fumarate (DMF) is an FDA-approved drug for the treatment of psoriasis and multiple sclerosis. DMF is known to stabilize the transcription factor Nrf2, which in turn induces the expression of antioxidant response element genes. It has also been shown that DMF influences autophagy and participates in the transcriptional control of inflammatory factors by inhibiting NF-κB and its downstream targets. DMF is receiving increasing attention for its potential to be repurposed for several diseases. This versatile molecule is indeed able to exert beneficial effects on different medical conditions through a pleiotropic mechanism, in virtue of its antioxidant, immunomodulatory, neuroprotective, anti-inflammatory, and anti-proliferative effects. A growing number of preclinical and clinical studies show that DMF may have important therapeutic implications for chronic diseases, such as cardiovascular and respiratory pathologies, cancer, eye disorders, neurodegenerative conditions, and systemic or organ specific inflammatory and immune-mediated diseases. This comprehensive review summarizes and highlights the plethora of DMF's beneficial effects and underlines its repurposing opportunities in a variety of clinical conditions.
Collapse
Affiliation(s)
- Giorgia Bresciani
- Section of Pharmacology, Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Federico Manai
- Department of Biology and Biotechnology L. Spallanzani, University of Pavia, Pavia, Italy
| | - Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology Vittorio Erspamer, Sapienza University, Rome, Italy
| | - Marialaura Amadio
- Section of Pharmacology, Department of Drug Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
6
|
Yan X, Li J, Wu D. The Role of Short-Chain Fatty Acids in Acute Pancreatitis. Molecules 2023; 28:4985. [PMID: 37446647 DOI: 10.3390/molecules28134985] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Acute pancreatitis (AP) is a digestive emergency and can develop into a systematic illness. The role of the gut in the progression and deterioration of AP has drawn much attention from researchers, and areas of interest include dysbiosis of the intestinal flora, weakened intestinal barrier function, and bacterial and endotoxin translocation. Short-chain fatty acids (SCFAs), as one of the metabolites of gut microbiota, have been proven to be depleted in AP patients. SCFAs help restore gut homeostasis by rebuilding gut flora, stabilizing the intestinal epithelial barrier, and regulating inflammation. SCFAs can also suppress systematic inflammatory responses, improve the injured pancreas, and prevent and protect other organ dysfunctions. Based on multiple beneficial effects, increasing SCFAs is an essential idea of gut protective treatment in AP. Specific strategies include the direct use of butyrate or indirect supplementation through fiber, pre/pro/synbiotics, or fecal microbiota transplantation as a promising adjective therapy to enteral nutrition.
Collapse
Affiliation(s)
- Xiaxiao Yan
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Eight-Year Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianing Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Dong Wu
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Clinical Epidemiology Unit, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
7
|
Yu Y, Li R, Pu L, Pan C, Guo C, Wang X. Citrus tangerine pith extract alleviates hypoxia-induced ileum damage in mice by modulating intestinal microbiota. Food Funct 2023. [PMID: 37314241 DOI: 10.1039/d3fo01096a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Visitors to high altitude are susceptible to hypoxia-induced acute intestinal mucosal barrier injury and severe gastrointestinal disorders, which are life-threatening. Citrus tangerine pith extract (CTPE) is rich in pectin and flavonoids and has been proved to enhance intestinal health and improve gut dysbiosis. In this study, we aim to explore the protective effect of CTPE on ileum injury induced by intermittent hypobaric hypoxia in a mouse model. Balb/c mice were divided into blank normoxia (BN), blank hypobaric hypoxia (BH), hypobaric hypoxia plus CTPE (TH), and hypobaric hypoxia plus Rhodiola extract (RH) groups. From the 6th day of gavage, mice in BH, TH, and RH groups were transferred into a hypobaric chamber at a simulated elevation of 6000 m for 8 hours per day for 10 days. Then half the mice were tested for small intestine movement, and others were used to evaluate intestinal physical barrier function, inflammation, and gut microbiota. Results showed that CTPE reversed the increase of intestinal peristalsis, effectively attenuated impaired structural integrity of ileum, improved the mRNA and protein expression levels of tight junction proteins, and reduced serum D-LA content in mice to alleviate hypoxia-induced mucosal barrier damage. Moreover, CTPE supplementation ameliorated hypoxia-induced intestinal inflammation response by significantly downregulating the proinflammatory cytokines IL-6, TNF-α and IFN-γ. By 16S rDNA gene sequencing of gut microbiota, CTPE significantly increased the abundance of probiotic Lactobacillus, suggesting that CTPE may be used as a prebiotic to regulate ecology of intestinal microorganisms. In addition, Spearman rank correlation analysis revealed that changed gut microbiota were significantly correlated with alteration of intestinal barrier function indexes. Taken together, these results indicate that CTPE effectively alleviates hypoxia-induced intestinal injury in mice and enhances intestinal integrity and barrier function by altering intestinal microbiota composition.
Collapse
Affiliation(s)
- Yijing Yu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Ran Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Lingling Pu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Cunyao Pan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Changjiang Guo
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Xinxing Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| |
Collapse
|
8
|
Manai F, Zanoletti L, Arfini D, Micco SGD, Gjyzeli A, Comincini S, Amadio M. Dimethyl Fumarate and Intestine: From Main Suspect to Potential Ally against Gut Disorders. Int J Mol Sci 2023; 24:9912. [PMID: 37373057 DOI: 10.3390/ijms24129912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Dimethyl fumarate (DMF) is a well-characterized molecule that exhibits immuno-modulatory, anti-inflammatory, and antioxidant properties and that is currently approved for the treatment of psoriasis and multiple sclerosis. Due to its Nrf2-dependent and independent mechanisms of action, DMF has a therapeutic potential much broader than expected. In this comprehensive review, we discuss the state-of-the-art and future perspectives regarding the potential repurposing of DMF in the context of chronic inflammatory diseases of the intestine, such as inflammatory bowel disorders (i.e., Crohn's disease and ulcerative colitis) and celiac disease. DMF's mechanisms of action, as well as an exhaustive analysis of the in vitro/in vivo evidence of its beneficial effects on the intestine and the gut microbiota, together with observational studies on multiple sclerosis patients, are here reported. Based on the collected evidence, we highlight the new potential applications of this molecule in the context of inflammatory and immune-mediated intestinal diseases.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Lisa Zanoletti
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
- Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Davide Arfini
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Simone Giorgio De Micco
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Arolda Gjyzeli
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Sergio Comincini
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Marialaura Amadio
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
9
|
Gut Microbiota Changes during Dimethyl Fumarate Treatment in Patients with Multiple Sclerosis. Int J Mol Sci 2023; 24:ijms24032720. [PMID: 36769041 PMCID: PMC9917003 DOI: 10.3390/ijms24032720] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The gut microbiota is involved in the development of the immune system and can modulate the risk for immune-mediated disorders such as multiple sclerosis (MS). Dysbiosis has been demonstrated in MS patients and its restoration by disease-modifying treatments (DMTs) is hypothesized. We aimed to study the changes in gut microbiota composition during the first 6 months of treatment with dimethyl fumarate (DMF), an oral DMT, and to identify the microorganisms associated with DMF side effects. We collected and analyzed the gut microbiota of 19 MS patients at baseline and after 1, 3, and 6 months of DMF treatment. We then cross-sectionally compared gut microbiota composition according to the presence of gastrointestinal (GI) symptoms and flushing. Overall, the gut microbiota biodiversity showed no changes over the 6-month follow-up. At the genus level, DMF was associated with decreased Clostridium abundance after 6 months. In subjects reporting side effects, a higher abundance of Streptococcus, Haemophilus, Clostridium, Lachnospira, Blautia, Subdoligranulum, and Tenericutes and lower of Bacteroidetes, Barnesiella, Odoribacter, Akkermansia, and some Proteobacteria families were detected. Our results suggest that gut microbiota may be involved in therapeutic action and side effects of DMF, representing a potential target for improving disease course and DMT tolerability.
Collapse
|
10
|
Dimethyl fumarate ameliorates fungal keratitis by limiting fungal growth and inhibiting pyroptosis. Int Immunopharmacol 2023; 115:109721. [PMID: 36641891 DOI: 10.1016/j.intimp.2023.109721] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/26/2022] [Accepted: 01/08/2023] [Indexed: 01/15/2023]
Abstract
PURPOSE We aimed to investigate the therapeutic role of dimethyl fumarate (DMF) in fungal keratitis. METHODS Human corneal epithelial cells (HCECs) and mouse models of fungal keratitis were used in this study. The antifungal effect of DMF on Aspergillus fumigatus (A. fumigatus) was confirmed by examining the minimum inhibitory concentration (MIC), biofilm formation, conidial adherence and corneal fungal loads. Slit-lamp photography, haematoxylin and eosin staining and immunostaining were used to assess the severity of corneal impairment. RT-PCR, western blot, ELISA, immunohistochemistry and immunostaining were performed to examine the effects of DMF on the expression of the inflammatory mediators during fungal infection. RESULTS In vitro, DMF limited A. fumigatus growth, biofilm formation, and conidial adherence and reduced the mRNA levels of AldA, GlkA, GAPDH, HxkA, PgkA, Sdh2, GelA and ChsF in A. fumigatus. In vivo, DMF effectively decreased corneal fungal loads. DMF attenuated corneal inflammatory impairment by suppressing inflammatory cell accumulation and downregulating cytokine expression. DMF notably downregulated the high expression of NLRP3, cleaved GSDMD, cleaved caspase-1, mature IL-1β and mature IL-18 induced by fungi. The production of Nrf2 and HO-1 could be further increased by DMF in infected HCECs. Nrf2 siRNA pretreatment counteracted DMF-mediated downregulation of the expression of the active forms of IL-18, IL-1β, caspase-1 and GSDMD. CONCLUSION DMF limits fungal growth by suppressing biofilm formation, conidial adherence and respiratory metabolism. It also exerts an anti-inflammatory effect on fungal keratitis by inhibiting pyroptosis, which could be regulated by Nrf2. Our results suggest that DMF plays a therapeutic role in fungal keratitis.
Collapse
|
11
|
Liu X, Tang H, Zhou Q, Zeng Y, Lu B, Chen D, Li Y, Qian J, Chen M, Zhao J, Xu Y, Wang M, Tan B. Gut microbiota composition in patients with advanced malignancies experiencing immune-related adverse events. Front Immunol 2023; 14:1109281. [PMID: 36891304 PMCID: PMC9986626 DOI: 10.3389/fimmu.2023.1109281] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/31/2023] [Indexed: 02/22/2023] Open
Abstract
Introduction The gut microbiota is implicated in the occurrence and severity of immune-related adverse events (irAEs), but the role it plays as well as its causal relationship with irAEs has yet to be established. Methods From May 2020 to August 2021, 93 fecal samples were prospectively collected from 37 patients with advanced thoracic cancers treated with anti-PD-1 therapy, and 61 samples were collected from 33 patients with various cancers developing different irAEs. 16S rDNA amplicon sequencing was performed. Antibiotic-treated mice underwent fecal microbiota transplantation (FMT) with samples from patients with and without colitic irAEs. Results Microbiota composition was significantly different in patients with and without irAEs (P=0.001) and with and without colitic-type irAEs (P=0.003). Bifidobacterium, Faecalibacterium, and Agathobacter were less abundant and Erysipelatoclostridium more abundant in irAE patients, while Bacteroides and Bifidobacterium were less abundant and Enterococcus more abundant in colitis-type irAE patients. Major butyrate-producing bacteria were also less abundant in patients with irAEs than those without (P=0.007) and in colitic vs. non-colitic irAE patients (P=0.018). An irAE prediction model had an AUC of 86.4% in training and 91.7% in testing. Immune-related colitis was more common in colitic-irAE-FMT (3/9) than non-irAE-FMT mice (0/9). Conclusions The gut microbiota is important in dictating irAE occurrence and type, especially for immune-related colitis, possibly by modulating metabolic pathways.
Collapse
Affiliation(s)
- Xinyu Liu
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China.,Eight-year Medical Doctor Program, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Hao Tang
- Department of Internal Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Qingyang Zhou
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Yanlin Zeng
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China.,School of Medicine, Tsinghua University, Beijing, China
| | - Bo Lu
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Dan Chen
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Yue Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Jiaming Qian
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Minjiang Chen
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Jing Zhao
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Bei Tan
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
12
|
Omarova MA, Rogovskii VS, Sadekov TS, Sadekova GI, Zhilenkova OG, Boyko AN. [Microbiota markers level in the blood and cerebrospinal fluid of patients with different types of multiple sclerosis and radiologically isolated syndrome]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:96-102. [PMID: 37560841 DOI: 10.17116/jnevro202312307296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
OBJECTIVE To assess the level of microbiota markers in the blood and cerebrospinal fluid (CSF) of patients with different types of multiple sclerosis (MS), people with radiologically isolated syndrome (RIS) and control subjects. MATERIAL AND METHODS We used gas chromatography-mass spectrometry (GC-MS) to evaluate the levels of microbiota markers in 69 patients with different types of MS (27 patients in the acute stage, 35 patients with MS in remission, 7 patients with primary-progressive MS), 10 people with RIS, and 47 control subjects (different diseases of the nervous system of a non-autoimmune or inflammatory nature). RESULTS We showed a statistically significant increase in the content of various microbiota markers in the CSF of patients with MS compared with the control group. We found no change in the content of these markers in blood of patients with MS. This suggests a change of markers of microbial load at the level of the central nervous system, but not at the level of the whole organism. The greatest number of statistically significant differences with the control group was found in the content of markers in CSF of patients with MS in remission. In the acute stage, on the contrary, we found no statistically significant differences compared to the control group. In particular, in CSF of patients with MS in remission, a statistically significant increase in the content of bacterial plasmalogen (4.5 times), and increase in the level of microbial markers specific to Peptostreptococcus anaerobius, Pseudomonas aeruginosa, Eubacterium, Bifidobacterium, Butirivibrio, Moraxella, Acinetobacter, Propionibacterium acnes, as well as an increase of markers of the Epstein-Barr virus were found. In addition, there was an increase of campesterol, the likely source of which is campesterol-producing microfungi. In the CSF of subjects with RIS there were a statistically significant increase in the level of markers of the Epstein-Barr virus, Propionibacterium acnes, as well as Pseudomonas, Moraxella, and Acinetobacter. CONCLUSION An association of MS with polymicrobial infection is possible. It is also likely that there is a certain pattern of increase of microbiota markers in the CSF of patients with MS, but not in blood.
Collapse
Affiliation(s)
- M A Omarova
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - V S Rogovskii
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - T Sh Sadekov
- Gabrichevsky Research Institute for Epidemiology and Microbiology, Moscow, Russia
| | - G I Sadekova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - O G Zhilenkova
- Gabrichevsky Research Institute for Epidemiology and Microbiology, Moscow, Russia
| | - A N Boyko
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
13
|
Jochems PGM, Heming B, Lapin D, Moonen NEL, Van den Ackerveken G, Masereeuw R. Bioengineered intestinal tubules as a tool to test intestinal biological efficacy of lettuce species. NPJ Sci Food 2022; 6:58. [PMID: 36513685 PMCID: PMC9747904 DOI: 10.1038/s41538-022-00175-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
Lettuce (Lactuca sativa) is one of the most consumed and cultivated vegetables globally. Its breeding is focused on the improvement of yield and disease resistance. However, potential detrimental or beneficial health effects for the consumer are often not targeted in the breeding programs. Here, a bioengineered intestinal tubule was used to assess the intestinal efficacy of extracts from five plant accessions belonging to four Lactuca species. These four species include the domesticated L. sativa, closely related wild species L. serriola, and phylogenetically more distant wild relatives L. saligna and L. virosa. We assessed the epithelial barrier integrity, cell viability, cell attachment, brush border enzyme activity, and immune markers. Extracts from L. sativa cv. Salinas decreased cell attachment and brush border enzyme activity. However, extracts from the non-edible wild species L. saligna and L. virosa reduced the epithelial barrier functions, cell attachment, cell viability, and brush border enzyme activity. Since wild species represent a valuable germplasm pool, the bioengineered intestinal tubules could open ways to evaluate the safety and nutritional properties of the lettuce breeding material originating from crosses with wild Lactuca species.
Collapse
Affiliation(s)
- Paulus G. M. Jochems
- grid.5477.10000000120346234Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Bo Heming
- grid.5477.10000000120346234Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Dmitry Lapin
- grid.5477.10000000120346234Translational Plant Biology, Department of Biology, Utrecht University, Utrecht, The Netherlands
| | | | - Guido Van den Ackerveken
- grid.5477.10000000120346234Translational Plant Biology, Department of Biology, Utrecht University, Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- grid.5477.10000000120346234Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
14
|
Diebold M, Meola M, Purushothaman S, Siewert LK, Pössnecker E, Roloff T, Lindberg RLP, Kuhle J, Kappos L, Derfuss T, Egli A, Pröbstel AK. Gut microbiota composition as a candidate risk factor for dimethyl fumarate-induced lymphopenia in multiple sclerosis. Gut Microbes 2022; 14:2147055. [PMID: 36398902 PMCID: PMC9677991 DOI: 10.1080/19490976.2022.2147055] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mounting evidence points towards a pivotal role of gut microbiota in multiple sclerosis (MS) pathophysiology. Yet, whether disease-modifying treatments alter microbiota composition and whether microbiota shape treatment response and side-effects remain unclear. In this prospective observational pilot study, we assessed the effect of dimethyl fumarate (DMF) on gut microbiota and on host/microbial metabolomics in a cohort of 20 MS patients. Combining state-of-the-art microbial sequencing, metabolome mass spectrometry, and computational analysis, we identified longitudinal changes in gut microbiota composition under DMF-treatment and an increase in citric acid cycle metabolites. Notably, DMF-induced lymphopenia, a clinically relevant safety concern, was correlated with distinct baseline microbiome signatures in MS patients. We identified gastrointestinal microbiota as a key therapeutic target for metabolic properties of DMF. By characterizing gut microbial composition as a candidate risk factor for DMF-induced lymphopenia, we provide novel insights into the role of microbiota in mediating clinical side-effects.
Collapse
Affiliation(s)
- Martin Diebold
- Departments of Neurology, Biomedicine and Clinical Research & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital and University of Basel, Basel, Switzerland,Institute of Neuropathology, Neurocenter, University Hospital Freiburg, University of Freiburg, Freiburg, Germany
| | - Marco Meola
- Division of Clinical Bacteriology and Mycology, University Hospital Basel, Basel, Switzerland,Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland,Institute for Medical Microbiology, University of Zurich, Zurich, Switzerland,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Srinithi Purushothaman
- Division of Clinical Bacteriology and Mycology, University Hospital Basel, Basel, Switzerland,Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland,Institute for Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Lena K Siewert
- Departments of Neurology, Biomedicine and Clinical Research & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Elisabeth Pössnecker
- Departments of Neurology, Biomedicine and Clinical Research & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Tim Roloff
- Division of Clinical Bacteriology and Mycology, University Hospital Basel, Basel, Switzerland,Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland,Institute for Medical Microbiology, University of Zurich, Zurich, Switzerland,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Raija LP Lindberg
- Departments of Neurology, Biomedicine and Clinical Research & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Departments of Neurology, Biomedicine and Clinical Research & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Ludwig Kappos
- Departments of Neurology, Biomedicine and Clinical Research & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Tobias Derfuss
- Departments of Neurology, Biomedicine and Clinical Research & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Adrian Egli
- Division of Clinical Bacteriology and Mycology, University Hospital Basel, Basel, Switzerland,Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland,Institute for Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Anne-Katrin Pröbstel
- Departments of Neurology, Biomedicine and Clinical Research & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital and University of Basel, Basel, Switzerland,CONTACT Anne-Katrin Pröbstel Departments of Neurology, Biomedicine and Clinical Research & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| |
Collapse
|
15
|
Yu HT, Zhang JQ, Sun MC, Chen H, Shi XM, You FP, Qiao SY. Polymeric Nanohybrids Engineered by Chitosan Nanoparticles and Antimicrobial Peptides as Novel Antimicrobials in Food Biopreservatives: Risk Assessment and Anti-Foodborne Pathogen Escherichia coli O157:H7 Infection by Immune Regulation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:12535-12549. [PMID: 36153996 DOI: 10.1021/acs.jafc.2c05308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Polymeric nanomaterials (APs) are gaining attention as promising clinical antimicrobials with rapidly increasing antibiotic resistance. Infections by zoonotic enterohemorrhagic Escherichia coli are a severe global threat to public health. Chitosan nanoparticles-microcin J25 (CNM), a class of APs engineered by bioactive peptides and chitosan nanoparticles, can be used as a novel antimicrobial agent against bacterial infections. However, the risk assessment of CNM on animal health or its potential immune modulation to treat serotype E. coli O157:H7 infection impacts in vivo are not well understood. Herein, our findings in mouse models uncovered that oral administration of low levels of CNM significantly increased the body weight and made beneficial effects on the lifespan or clinical signs, accompanied by a significant improvement in gut health, including enhancing the intestinal barrier, immune modulation, and changes in gut microbiota compositions or metabolites. However, high concentrations of CNM induced serious adverse effects, negatively improving intestinal health targets. Anti-infective results proved that oral 0.1% CNM enhances host defense against E. coli O157:H7 infection by improving immune functions and modulating the Th1/Th2 balance. In summary, these findings uncover an instrumental link between the dosage and toxicity risk, suggesting that APs need to be comprehensively assessed for risk before application as safe and reliable food preservatives or therapeutic agents. In addition, CNM as a promising AP may markedly enhance host immunity and therapeutic effects by oral administration.
Collapse
Affiliation(s)
- Hai-Tao Yu
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Jia-Qi Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Ming-Chao Sun
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Han Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Xiu-Mei Shi
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Fu-Ping You
- Institute of Systems Biomedicine, Department of Immunology, School of Basic Medical Sciences, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, P. R. China
| | - Shi-Yan Qiao
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| |
Collapse
|
16
|
Li L, Wang M, Bao J, Wang N, Huang Y, He S, Chen B, Yan F. Periodontitis may impair the homeostasis of systemic bone through regulation of gut microbiota in
ApoE
‐/‐
mice. J Clin Periodontol 2022; 49:1304-1319. [PMID: 35871602 DOI: 10.1111/jcpe.13708] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/01/2022] [Accepted: 07/21/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Lili Li
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Min Wang
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Jun Bao
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Nannan Wang
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Yuezhen Huang
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Shasha He
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Bin Chen
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| | - Fuhua Yan
- Department of Periodontology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
- Central laboratory of Stomatology, Nangjing Stomatological Hospital Medical School of Nanjing University Nanjing Jiangsu P.R. China
| |
Collapse
|
17
|
Dey DK, Kang JI, Bajpai VK, Kim K, Lee H, Sonwal S, Simal-Gandara J, Xiao J, Ali S, Huh YS, Han YK, Shukla S. Mycotoxins in food and feed: toxicity, preventive challenges, and advanced detection techniques for associated diseases. Crit Rev Food Sci Nutr 2022; 63:8489-8510. [PMID: 35445609 DOI: 10.1080/10408398.2022.2059650] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mycotoxins are produced primarily as secondary fungal metabolites. Mycotoxins are toxic in nature and naturally produced by various species of fungi, which usually contaminate food and feed ingredients. The growth of these harmful fungi depends on several environmental factors, such as pH, humidity, and temperature; therefore, the mycotoxin distribution also varies among global geographical areas. Various rules and regulations regarding mycotoxins are imposed by the government bodies of each country, which are responsible for addressing global food and health security concerns. Despite this legislation, the incidence of mycotoxin contamination is continuously increasing. In this review, we discuss the geographical regulatory guidelines and recommendations that are implemented around the world to control mycotoxin contamination of food and feed products. Researchers and inventors from various parts of the world have reported several innovations for controlling mycotoxin-associated health consequences. Unfortunately, most of these techniques are restricted to laboratory scales and cannot reach users. Consequently, to date, no single device has been commercialized that can detect all mycotoxins that are naturally available in the environment. Therefore, in this study, we describe severe health hazards that are associated with mycotoxin exposure, their molecular signaling pathways and processes of toxicity, and their genotoxic and cytotoxic effects toward humans and animals. We also discuss recent developments in the construction of a sensitive and specific device that effectively implements mycotoxin identification and detection methods. In addition, our study comprehensively examines the recent advancements in the field for mitigating the health consequences and links them with the molecular and signaling pathways that are activated upon mycotoxin exposure.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Gyeongsan, Republic of Korea
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ji In Kang
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon, Republic of Korea
| | - Vivek K Bajpai
- Department of Energy and Materials Engineering, Dongguk University, Seoul, Republic of Korea
| | - Kwanwoo Kim
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, Republic of Korea
| | - Hoomin Lee
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, Republic of Korea
| | - Sonam Sonwal
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, Republic of Korea
| | - Jesus Simal-Gandara
- Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, Ourense, Spain
| | - Jianbo Xiao
- Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, Ourense, Spain
| | - Sajad Ali
- Department of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Yun Suk Huh
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, Republic of Korea
| | - Yong-Kyu Han
- Department of Energy and Materials Engineering, Dongguk University, Seoul, Republic of Korea
| | - Shruti Shukla
- TERI-Deakin Nanobiotechnology Centre, The Energy and Resources Institute, Gurugram, Haryana, India
| |
Collapse
|
18
|
Xiong Y, Ji L, Zhao Y, Liu A, Wu D, Qian J. Sodium Butyrate Attenuates Taurocholate-Induced Acute Pancreatitis by Maintaining Colonic Barrier and Regulating Gut Microorganisms in Mice. Front Physiol 2022; 13:813735. [PMID: 35370779 PMCID: PMC8969109 DOI: 10.3389/fphys.2022.813735] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 02/09/2022] [Indexed: 12/12/2022] Open
Abstract
BackgroundAcute pancreatitis (AP) damages the intestinal barrier, which aggravates AP. Butyrate exhibits anti-inflammatory effects in AP, but it is unknown if such a protective effect is associated with the regulation of gut microorganisms. We aim to investigate the effects of sodium butyrate (SB) on pancreatic inflammation, colonic barrier, and gut microorganisms.MethodsC57BL/6 mice were divided into groups of sham operation (Sham), AP, 200 mg/kg SB intervention (SB-200), and 500 mg/kg SB intervention group (SB-500). Samples were harvested 24 h after the model was established. The gut microbiota was analyzed using 16S rRNA gene sequencing.ResultsPancreatic infiltration of neutrophils, macrophages, and M2-type macrophages was significantly reduced in the SB-500 intervention group. Supplementation of SB-500 improved colon mucosal histology and the expression of ZO-1 and occluding. The relative abundance of Alloprevotella and Muribaculaceae was increased and that of Akkermansia was decreased in the SB-500 group compared with the AP group. Ruminococcaceae was the most significantly increased species and Prevotellaceae was the most significantly decreased species in the SB-500 group compared with the AP group.ConclusionHigh dose of SB inhibits pancreatic inflammation probably by maintaining the intestinal barrier and regulating gut microbiota in mice with AP.
Collapse
|
19
|
The Defect in Regulatory T Cells in Psoriasis and Therapeutic Approaches. J Clin Med 2021; 10:jcm10173880. [PMID: 34501328 PMCID: PMC8432197 DOI: 10.3390/jcm10173880] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by accelerated tumor necrosis factor-α/interleukin (IL)-23/IL-17 axis. Patients with psoriasis manifest functional defects in CD4+CD25+ forkhead box protein 3 (Foxp3)+ regulatory T cells (Tregs), which suppress the excess immune response and mediate homeostasis. Defects in Tregs contribute to the pathogenesis of psoriasis and may attribute to enhanced inhibition and/or impaired stimulation of Tregs. IL-23 induces the conversion of Tregs into type 17 helper T (Th17) cells. IL-17A reduces transforming growth factor (TGF)-β1 production, Foxp3 expression, and suppresses Treg activity. Short-chain fatty acids (SCFAs), butyrate, propionate, and acetate are microbiota-derived fermentation products that promote Treg development and function by inducing Foxp3 expression or inducing dendritic cells or intestinal epithelial cells to produce retinoic acids or TGF-β1, respectively. The gut microbiome of patients with psoriasis revealed reduced SCFA-producing bacteria, Bacteroidetes, and Faecallibacterium, which may contribute to the defect in Tregs. Therapeutic agents currently used, viz., anti-IL-23p19 or anti-IL-17A antibodies, retinoids, vitamin D3, dimethyl fumarate, narrow-band ultraviolet B, or those under development for psoriasis, viz., signal transducer and activator of transcription 3 inhibitors, butyrate, histone deacetylase inhibitors, and probiotics/prebiotics restore the defected Tregs. Thus, restoration of Tregs is a promising therapeutic target for psoriasis.
Collapse
|
20
|
Ghorbani Y, Schwenger KJP, Allard JP. Manipulation of intestinal microbiome as potential treatment for insulin resistance and type 2 diabetes. Eur J Nutr 2021; 60:2361-2379. [PMID: 33651137 DOI: 10.1007/s00394-021-02520-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/16/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE Increasing evidence suggests that the intestinal microbiome (IM) and bacterial metabolites may influence glucose homeostasis, energy expenditure and the intestinal barrier integrity and lead to the presence of systemic low-grade inflammation, all of which can contribute to insulin resistance (IR) and type 2 diabetes (T2D). The purpose of this review is to explore the role of the IM and bacterial metabolites in the pathogenesis and treatment of these conditions. RESULTS This review summarizes research focused on how to modulate the IM through diet, prebiotics, probiotics, synbiotics and fecal microbiota transplant in order to treat IR and T2D. CONCLUSION There is an abundance of evidence suggesting a role for IM in the pathogenesis of IR and T2D based on reviewed studies using various methods to modulate IM and metabolites. However, the results are inconsistent. Future research should further assess this relationship.
Collapse
Affiliation(s)
- Yasaman Ghorbani
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital, University Health Network, Toronto, Canada
| | | | - Johane P Allard
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Toronto General Hospital, University Health Network, Toronto, Canada.
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada.
- Department of Medicine, University of Toronto, Toronto, Canada.
- Department of Medicine, Division of Gastroenterology, Toronto General Hospital, 585 University Avenue, 9N-973, Toronto, ON, M5G 2N2, Canada.
| |
Collapse
|
21
|
Influence of immunomodulatory drugs on the gut microbiota. Transl Res 2021; 233:144-161. [PMID: 33515779 PMCID: PMC8184576 DOI: 10.1016/j.trsl.2021.01.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/10/2020] [Accepted: 01/26/2021] [Indexed: 12/17/2022]
Abstract
Immunomodulatory medications are a mainstay of treatment for autoimmune diseases and malignancies. In addition to their direct effects on immune cells, these medications also impact the gut microbiota. Drug-induced shifts in commensal microbes can lead to indirect but important changes in the immune response. We performed a comprehensive literature search focusing on immunotherapy/microbe interactions. Immunotherapies were categorized into 5 subtypes based on their mechanisms of action: cell trafficking inhibitors, immune checkpoint inhibitors, immunomodulators, antiproliferative drugs, and inflammatory cytokine inhibitors. Although no consistent relationships were observed between types of immunotherapy and microbiota, most immunotherapies were associated with shifts in specific colonizing bacterial taxa. The relationships between colonizing microbes and drug efficacy were not well-studied for autoimmune diseases. In contrast, the efficacy of immune checkpoint inhibitors for cancer was tied to the baseline composition of the gut microbiota. There was a paucity of high-quality data; existing data were generated using heterogeneous sampling and analytic techniques, and most studies involved small numbers of participants. Further work is needed to elucidate the extent and clinical significance of immunotherapy effects on the human microbiome.
Collapse
|
22
|
Shi T, Bian X, Yao Z, Wang Y, Gao W, Guo C. Quercetin improves gut dysbiosis in antibiotic-treated mice. Food Funct 2021; 11:8003-8013. [PMID: 32845255 DOI: 10.1039/d0fo01439g] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The diversity and activity of the gut microbiota residing in humans and animals are significantly influenced by the diet. Quercetin, one of the representative polyphenols in human diets, possesses a wide range of biological properties. The aim of this study was to investigate the prebiotic effects of quercetin in antibiotic-treated mice. Gut dysbiosis was successfully induced in mice by treatment with an antibiotic cocktail. Gas chromatography and 16S rDNA high-throughput sequencing techniques were used to investigate short-chain fatty acid content and gut microbial diversity and composition. The results showed that quercetin supplementation significantly improved the diversity of the gut bacterial community in antibiotic-treated mice (P < 0.05). Meanwhile, intestinal barrier function was also recovered remarkably as indicated by a decrease in the content of serum d-lactic acid and the activity of serum diamine oxidase (P < 0.05). The length of intestinal villi and mucosal thickness were also significantly increased in response to quercetin treatment (P < 0.05). Furthermore, the production of butyrate in faeces was enhanced significantly in quercetin-treated mice (P < 0.05). In conclusion, quercetin is effective in recovering gut microbiota in mice after antibiotic treatment and may act as a prebiotic in combatting gut dysbiosis.
Collapse
Affiliation(s)
- Tala Shi
- Institute of Environmental and Operational Medicine, Tianjin, China. and Department of Nutrition and Food Hygiene, Binzhou Medical University, Yantai, China
| | - Xiangyu Bian
- Institute of Environmental and Operational Medicine, Tianjin, China.
| | - Zhanxin Yao
- Institute of Environmental and Operational Medicine, Tianjin, China.
| | - Yawen Wang
- Institute of Environmental and Operational Medicine, Tianjin, China.
| | - Weina Gao
- Institute of Environmental and Operational Medicine, Tianjin, China.
| | - Changjiang Guo
- Institute of Environmental and Operational Medicine, Tianjin, China.
| |
Collapse
|
23
|
Liu H, Wang C, Gu X, Zhao J, Nie C, Zhang W, Ma X. Dietary Montmorillonite Improves the Intestinal Mucosal Barrier and Optimizes the Intestinal Microbial Community of Weaned Piglets. Front Microbiol 2020; 11:593056. [PMID: 33324372 PMCID: PMC7723851 DOI: 10.3389/fmicb.2020.593056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/13/2020] [Indexed: 01/22/2023] Open
Abstract
The study investigated the impact of dietary montmorillonite on the growth performance, intestinal mucosal barrier, and microbial community in weaned piglets with control group (CON) and dietary supplementation of 0.2% montmorillonite (0.2% M). Compared with the CON group, 0.2% M feed in the diet increased the average daily gain (ADG) on days 15-35 and day 1-35 and the average daily feed intake on days 1-35 (ADFI) (0.05 < P < 0.1). Besides, higher villus height of the duodenum and jejunum and lower crypt depth of duodenum and colon were revealed in the 0.2% M group than in the CON group (P < 0.05). Moreover, the V/C (ratio of the villus height and crypt depth) in the 0.2% M group was increased compared to that in the CON group both from the duodenum and ileum (P < 0.05). The relative mRNA expression of mucin-1, ITGB1 (β1-integrins), and PKC (protein kinase C) of ileum in the 0.2% M group were upregulated (P < 0.05) compared to that in the CON group. The digesta sample of ileum from piglets in the 0.2% M group contained greater (P < 0.05) intestinal bacterial diversity and abundances of probiotics, such as Streptococcus, Eubacterium_rectale_group, and Lactobacillus, which could promote the synthesis of carbon-containing biomolecules. Overall, dietary supplementation of 0.2% M was shown to have a tendency to improve the growth performance of weaned piglets and may enhance their intestinal mucosal barrier function via altering the gut microbiota.
Collapse
Affiliation(s)
- Han Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Congmin Wang
- Department of Dermatology, Seventh Medical Center of Chinese PLA (People’s Liberation Army) General Hospital, Beijing, China
| | - Xueling Gu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jing Zhao
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Cunxi Nie
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Wenju Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| |
Collapse
|
24
|
Wei C, Yu L, Qiao N, Zhao J, Zhang H, Zhai Q, Tian F, Chen W. Progress in the distribution, toxicity, control, and detoxification of patulin: A review. Toxicon 2020; 184:83-93. [DOI: 10.1016/j.toxicon.2020.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/24/2020] [Accepted: 05/15/2020] [Indexed: 01/09/2023]
|
25
|
Xiong X, Liang J, Xu Y, Liu J, Liu Y. The wound healing effects of the Tilapia collagen peptide mixture TY001 in streptozotocin diabetic mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:2848-2858. [PMID: 31646634 DOI: 10.1002/jsfa.10104] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 09/12/2019] [Accepted: 09/18/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND The Tilapia collagen peptides mixture TY001 is effective in promoting wound healing in acetic acid-induced skin lesions in zebrafish and in protecting against lipopolysaccharide-induced inflammation and disruption of glucose metabolism in mice. The present study aimed to further examine the wound healing effects of TY001 in streptozotocin-induced diabetic mice. METHODS Full-thickness skin excision wounds were created with 8-mm biopsy punches and TY001 was administered via drinking water (15, 30 and 45 g L-1 in emulsion) for 15 days. RESULTS Wound healing was delayed in diabetic mice but was promoted by TY001 after 5, 10 or 15 days of treatment. Collagen deposition and tissue hydroxyproline contents were increased by TY001. The expressions of insulin growth factor-1, basic fibroblast growth factor, platelet-derived growth factor, transforming growth facts β1, vascular endothelial growth factor and epidermal growth factor were increased by TY001, as indicated by immunobiochemistry and a quantitative polymerase chain reaction. Diabetes-associated serum pro-inflammatory cytokines interleukin (IL)-1β and IL-8 were decreased, whereas anti-inflammatory IL-10 and nitric oxide were increased by TY001, along with increased tissue antioxidant superoxide dismutase and catalase activities. Diabetes-reduced serum protein levels were also recovered by TY001 CONCLUSION: Taken together, Tilapia collagen peptide mixture TY001 was effective with respect to enhancing diabetes-associated wound healing delay, probably via increasing growth factors and collagen deposition in the wound, attenuating diabetes-induced prolonged inflammation, increasing tissue antioxidants and providing nutritional support in diabetic mice. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaoyun Xiong
- Yabao Pharmaceutical Group Co., Ltd, Fenglingdu, China
| | - Jun Liang
- Yabao Pharmaceutical Group Co., Ltd, Fenglingdu, China
| | - Yiqiao Xu
- Hunter Biotechnology, Inc., Hangzhou, China
| | - Jie Liu
- Zunyi Medical University, Zunyi, China
| | - Yi Liu
- The Center for Disease Control and Prevention of Shaanxi Province, Xi'an, China
| |
Collapse
|
26
|
Microbiome in Multiple Sclerosis; Where Are We, What We Know and Do Not Know. Brain Sci 2020; 10:brainsci10040234. [PMID: 32295236 PMCID: PMC7226078 DOI: 10.3390/brainsci10040234] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023] Open
Abstract
An increase of multiple sclerosis (MS) incidence has been reported during the last decade, and this may be connected to environmental factors. This review article aims to encapsulate the current advances targeting the study of the gut-brain axis, which mediates the communication between the central nervous system and the gut microbiome. Clinical data arising from many research studies, which have assessed the effects of administered disease-modifying treatments in MS patients to the gut microbiome, are also recapitulated.
Collapse
|
27
|
Zhao Z, Cai W, Song L, Mu X, Hu Y. Comprehensive Property Investigation of Mold Inhibitor Treated Raw Cotton and Ramie Fabric. MATERIALS 2020; 13:ma13051105. [PMID: 32121662 PMCID: PMC7085025 DOI: 10.3390/ma13051105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/10/2020] [Accepted: 02/27/2020] [Indexed: 12/17/2022]
Abstract
At present, research rarely focuses on side effects of the use of mold inhibitors on raw cotton and ramie fabric. Four different mold inhibitors (dimethyl fumarate (DMF), ethyl p-hydroxybenzoate (EHB), propyl p-hydroxybenzoate (PHB), and calcium sorbate (CS)) were used to treat raw cotton and ramie fabric through a dipping method. The optical properties, wettability, thermal conductivity, thermal stability, and combustion properties of treated cotton and ramie samples have been investigated. The reflectance of UV light was improved by the addition of mold inhibitors. In addition, the presence of EHB, PHB, and CS improved the wettability of raw cotton and ramie fabric. It was found that thermal conductivity was slightly increased, influencing the heat insulation effect of the fabrics. Since the additives are flammable, the presence of DMF, EHB, and PHB caused an increase in pHRR and THR for combustion of cotton samples. This addition of CS caused a decrease in pHRR and THR of cotton due to the flame retardancy of CS. This comprehensive investigation of the properties of raw cotton and ramie fabrics treated with these materials should provide a basis for the choice of mold inhibitors.
Collapse
Affiliation(s)
- Zhixin Zhao
- State Key Laboratory of Fire Science, University of Science and Technology of China, Hefei 230026, China; (W.C.); (L.S.)
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, China;
| | - Wei Cai
- State Key Laboratory of Fire Science, University of Science and Technology of China, Hefei 230026, China; (W.C.); (L.S.)
| | - Lei Song
- State Key Laboratory of Fire Science, University of Science and Technology of China, Hefei 230026, China; (W.C.); (L.S.)
| | - Xiaowei Mu
- State Key Laboratory of Fire Science, University of Science and Technology of China, Hefei 230026, China; (W.C.); (L.S.)
- Correspondence: (X.M.); (Y.H.)
| | - Yuan Hu
- State Key Laboratory of Fire Science, University of Science and Technology of China, Hefei 230026, China; (W.C.); (L.S.)
- Correspondence: (X.M.); (Y.H.)
| |
Collapse
|
28
|
Pitarokoili K, Bachir H, Sgodzai M, Grüter T, Haupeltshofer S, Duscha A, Pedreiturria X, Motte J, Gold R. Induction of Regulatory Properties in the Intestinal Immune System by Dimethyl Fumarate in Lewis Rat Experimental Autoimmune Neuritis. Front Immunol 2019; 10:2132. [PMID: 31552056 PMCID: PMC6746892 DOI: 10.3389/fimmu.2019.02132] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 08/23/2019] [Indexed: 12/22/2022] Open
Abstract
Objective: Dimethyl fumarate (DMF) exerts immunomodulatory and neuroprotective effects in the animal model of experimental autoimmune neuritis (EAN) in the Lewis rat. DMF has been shown to modulate gut microbiota in veterinary medicine, however the effects of oral DMF on the gut-associated lymphoid tissue (GALT) remain unknown. Methods: Lewis rats were treated orally twice daily with DMF up to day 10 after immunization with immunogenic P2 peptide. Histological, flow cytometric and RT-PCR analyses of the GALT (intraepithelial layer, lamina propria, and Peyer patches) in duodenum, jejunum, and ileum were performed ex vivo. Moreover, cell transfer experiments were used to examine the protective effects of GALT regulatory T cells of the Peyer patches. Results: In the upper layers of duodenum, DMF induced a reduction of the toll-like receptor 4 (TLR4) mRNA expression. This was combined by a decrease of the pro-inflammatory lamina propria IFN-γ mRNA expression. In the ileum, we detected an immunoregulatory phenotype characterized by an increase of FoxP3 mRNA expression and of the nuclear factor (erythroid-derived-2)- like 2 (Nrf2) downstream molecule heme oxygenase-1 (HO-1) mRNA. Finally, CD4+ CD25+ regulatory T cells were increased in the Peyer patches. In vivo, the protective effect of these regulatory cells was verified by cell transfer into recipient EAN rats. Conclusions: Our results identified a novel immunomodulatory effect of DMF through the different regions and layers of the small intestine, which led to an increase of regulatory T cells, exerting a protective role in experimental neuritis.
Collapse
Affiliation(s)
- Kalliopi Pitarokoili
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Hussein Bachir
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Melissa Sgodzai
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Thomas Grüter
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Steffen Haupeltshofer
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Alexander Duscha
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Xiomara Pedreiturria
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Jeremias Motte
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
29
|
Camara-Lemarroy CR, Metz L, Meddings JB, Sharkey KA, Wee Yong V. The intestinal barrier in multiple sclerosis: implications for pathophysiology and therapeutics. Brain 2019; 141:1900-1916. [PMID: 29860380 DOI: 10.1093/brain/awy131] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/24/2018] [Indexed: 12/12/2022] Open
Abstract
Biological barriers are essential for the maintenance of homeostasis in health and disease. Breakdown of the intestinal barrier is an essential aspect of the pathophysiology of gastrointestinal inflammatory diseases, such as inflammatory bowel disease. A wealth of recent studies has shown that the intestinal microbiome, part of the brain-gut axis, could play a role in the pathophysiology of multiple sclerosis. However, an essential component of this axis, the intestinal barrier, has received much less attention. In this review, we describe the intestinal barrier as the physical and functional zone of interaction between the luminal microbiome and the host. Besides its essential role in the regulation of homeostatic processes, the intestinal barrier contains the gut mucosal immune system, a guardian of the integrity of the intestinal tract and the whole organism. Gastrointestinal disorders with intestinal barrier breakdown show evidence of CNS demyelination, and content of the intestinal microbiome entering into the circulation can impact the functions of CNS microglia. We highlight currently available studies suggesting that there is intestinal barrier dysfunction in multiple sclerosis. Finally, we address the mechanisms by which commonly used disease-modifying drugs in multiple sclerosis could alter the intestinal barrier and the microbiome, and we discuss the potential of barrier-stabilizing strategies, including probiotics and stabilization of tight junctions, as novel therapeutic avenues in multiple sclerosis.
Collapse
Affiliation(s)
- Carlos R Camara-Lemarroy
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Luanne Metz
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jonathan B Meddings
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keith A Sharkey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
30
|
Food Supplements to Mitigate Detrimental Effects of Pelvic Radiotherapy. Microorganisms 2019; 7:microorganisms7040097. [PMID: 30987157 PMCID: PMC6518429 DOI: 10.3390/microorganisms7040097] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/21/2019] [Accepted: 03/28/2019] [Indexed: 12/12/2022] Open
Abstract
Pelvic radiotherapy has been frequently reported to cause acute and late onset gastrointestinal (GI) toxicities associated with significant morbidity and mortality. Although the underlying mechanisms of pelvic radiation-induced GI toxicity are poorly understood, they are known to involve a complex interplay between all cell types comprising the intestinal wall. Furthermore, increasing evidence states that the human gut microbiome plays a role in the development of radiation-induced health damaging effects. Gut microbial dysbiosis leads to diarrhea and fatigue in half of the patients. As a result, reinforcement of the microbiome has become a hot topic in various medical disciplines. To counteract GI radiotoxicities, apart from traditional pharmacological compounds, adjuvant therapies are being developed including food supplements like vitamins, prebiotics, and probiotics. Despite the easy, cheap, safe, and feasible approach to protect patients against acute radiation-induced toxicity, clinical trials have yielded contradictory results. In this review, a detailed overview is given of the various clinical, intestinal manifestations after pelvic irradiation as well as the role of the gut microbiome herein. Furthermore, whilst discussing possible strategies to prevent these symptoms, food supplements are presented as auspicious, prophylactic, and therapeutic options to mitigate acute pelvic radiation-induced GI injury by exploring their molecular mechanisms of action.
Collapse
|
31
|
Gut microbiota dysbiosis worsens the severity of acute pancreatitis in patients and mice. J Gastroenterol 2019; 54:347-358. [PMID: 30519748 DOI: 10.1007/s00535-018-1529-0] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 11/08/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND The gut is implicated in the pathogenesis of acute pancreatitis (AP) and the infectious complications of AP are commonly associated with enteric bacteria, yet whether gut microbiota dysbiosis participants in AP severity remains largely unknown. METHODS We collected clinical information and fecal samples from 165 adult participants, including 41 with mild AP (MAP), 59 with moderately severe AP (MSAP), 30 with severe AP (SAP) and 35 healthy controls (HC). The serum inflammatory cytokines and gut barrier indexes were detected. Male C57BL/6 mice with AP were established and injuries of pancreas were evaluated in antibiotic-treated mice, germ-free mice as well as those transplanted with fecal microbiota. The gut microbiota was analyzed by 16S rRNA gene sequencing. RESULTS The structure of gut microbiota was significantly different between AP and HC, and the disturbed microbiota was closely correlated with systematic inflammation and gut barrier dysfunction. Notably, the microbial composition changed further with the worsening of AP and the abundance of beneficial bacteria such as Blautia was decreased in SAP compared with MAP and MSAP. The increased capacity for the inferred pathway, bacterial invasion of epithelial cells in AP, highly correlated with the abundance of Escherichia-Shigella. Furthermore, the antibiotic-treated mice and germ-free mice exhibited alleviated pancreatic injury after AP induction and subsequent fecal microbiota transplantation in turn exacerbated the disease. CONCLUSIONS This study identifies the gut microbiota as an important mediator during AP and its dysbiosis is associated with AP severity, which suggests its role as potential therapeutic target.
Collapse
|
32
|
Wang D, Yan S, Yan J, Teng M, Meng Z, Li R, Zhou Z, Zhu W. Effects of triphenyl phosphate exposure during fetal development on obesity and metabolic dysfunctions in adult mice: Impaired lipid metabolism and intestinal dysbiosis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 246:630-638. [PMID: 30605818 DOI: 10.1016/j.envpol.2018.12.053] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 06/09/2023]
Abstract
Previous in vitro studies have implied that triphenyl phosphate (TPHP) may act as an obesogen. However, its specific contributions to the progression of obesity and related metabolic diseases are still unclear in vivo in mice. In this study, we evaluated the effects of in utero and lactational exposure to three doses of TPHP (10, 100, and 1000 μg/kg BW) on obesity and metabolic dysfunctions in adult male mice fed a low-fat diet (LFD) or high-fat diet (HFD), by examining body weight, liver weight, histopathology, blood biochemistry, gene expression, and gut microbiota compositions and metabolic functions. Results showed that TPHP exposure led to increased body weight, liver weight, fat mass, hepatic steatosis, impaired glucose homeostasis, and insulin resistance, and mRNA levels of genes involved in lipid metabolism, especially lipogenesis and lipid accumulation, were significantly altered by TPHP treatment. Gas chromatography-mass spectrometry (GC-MS) analysis further supported the changes in fatty acid composition. Intestinal flora measurements by 16S rRNA gene sequencing and 1H NMR based fecal metabolomics indicated that TPHP treatment modulated gut microbiome composition and influenced host-gut co-metabolism, especially for bile acids and short chain fatty acids (SCFAs). These results suggest that fetal exposure to TPHP can promote the development of obesity and metabolic dysfunctions in adult mice.
Collapse
Affiliation(s)
- Dezhen Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, PR China
| | - Sen Yan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, PR China
| | - Jin Yan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, PR China
| | - Miaomiao Teng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, PR China
| | - Zhiyuan Meng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, PR China
| | - Ruisheng Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, PR China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, PR China
| | - Wentao Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, PR China.
| |
Collapse
|
33
|
Xiao L, Chen B, Feng D, Yang T, Li T, Chen J. TLR4 May Be Involved in the Regulation of Colonic Mucosal Microbiota by Vitamin A. Front Microbiol 2019; 10:268. [PMID: 30873131 PMCID: PMC6401601 DOI: 10.3389/fmicb.2019.00268] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/01/2019] [Indexed: 12/14/2022] Open
Abstract
Objectives: To investigate the specific role of Toll-like receptor 4 (TLR4) in the regulation of the intestinal mucosa-associated microbiota by vitamin A (VA). Methods: Both TLR4-/- (knockout, KO) and wild-type (WT) female mice were randomly fed a VA normal (VAN) or VA deficient (VAD) diet for 4 weeks to establish the following four mouse model groups: TLR4-/- mice fed a VAN diet (KO VAN), TLR4-/- mice fed a VAD diet (KO VAD), WT mice fed a VAN diet (WT VAN), and WT mice fed a VAD diet (WT VAD). Then, the mice from each experimental group were mated with male mice with the same genetic background. The pups in the KO VAD and WT VAD groups were subsequently fed the VAD diet after weaning, while the pups in the KO VAN and WT VAN groups were fed the VAN diet continuously after weaning. The serum retinol levels of 7-week-old offspring were determined using high-performance liquid chromatography, and colons were collected from mice in each group and analyzed via 16S rRNA gene sequencing using an Illumina MiSeq platform to characterize the overall microbiota of the samples. Results: The abundance and evenness of the colon mucosa-associated microbiota were unaffected by dietary VA and TLR4 KO. VAD decreased the abundance of Anaerotruncus (Firmicutes), Oscillibacter (Firmicutes), Lachnospiraceae _NK4A136 _group (Firmicutes) and Mucispirillum (Deferribacteres) and increased the abundance of Parasutterella (Proteobacteria). TLR4 KO decreased the abundance of Bacteroides (Bacteroidetes) and Alloprevotella (Bacteroidetes). However, the abundance of Allobaculum (Firmicutes), Ruminiclostridium_9 (Firmicutes), Alistipes (Bacteroidetes), and Rikenellaceae_RC9 (Bacteroidetes) impacted the interaction between VA and TLR4. Conclusion: TLR4 may play a pivotal role in regulation of the intestinal mucosa-associated microbiota by VA to maintain the intestinal microecology.
Collapse
Affiliation(s)
- Lu Xiao
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Baolin Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Di Feng
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Ting Yang
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Tingyu Li
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Jie Chen
- Children's Nutrition Research Center, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| |
Collapse
|
34
|
Younan S, Sakita GZ, Coluna JGY, Rufino MN, Keller R, Bremer-Neto H. Probiotic mitigates the toxic effects of potassium dichromate in a preclinical study: a randomized controlled trial. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2019; 99:183-190. [PMID: 29851070 DOI: 10.1002/jsfa.9159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/19/2018] [Accepted: 05/28/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND The present study aimed to evaluate the nutritional, physiological and biochemical effects of dietary supplementation of an association of probiotic bacteria in rats intoxicated with chromium (VI). Ninety-six male rats, recently weaned, were randomly divided into eight groups (n = 12): Control, DK12, DK24 and DK36 (0, 0.12, 0.24 and 0.36 g kg-1 of K2 Cr2 O7 incorporated in the basal feed, respectively) and groups Prob, DK12 + Prob, DK24 + Prob and DK36 + Prob received a progressive dose of 0, 0.12, 0.24 and 0.36 g kg-1 of K2 Cr2 O7 incorporated in the basal feed and supplemented with 0.02 g kg-1 of an association of probiotic bacteria (Lactobacillus acidophilus, Enterococcus faecium, Bifidobacterium thermophilum and Bifidobacterium longum). RESULTS After 90 days, we observed significant (P < 0.05) and dose-dependent alterations from incorporation of increasing doses of chromium (VI) related to nutritional, physiological and biochemical parameters. These changes were attenuated (P < 0.05) with probiotic supplementation. CONCLUSION Supplementation with probiotics in the diet beneficially modified the nutritional and physiological parameters, as well as hepatic, renal, glycemic and lipid profiles, of animals intoxicated with increasing doses of K2 Cr2 O7 . © 2018 Society of Chemical Industry.
Collapse
Affiliation(s)
- Soraia Younan
- Department of Functional Sciences, Universidade do Oeste Paulista, Presidente Prudente, Brazil
| | - Gabriel Zanuto Sakita
- Animal Nutrition Laboratory, Universidade de São Paulo, Centro de Energia Nuclear na Agricultura, Piracicaba, Brazil
| | - João Gabriel Younan Coluna
- Faculty of Medicine, Department of Functional Sciences, Universidade do Oeste Paulista, Presidente Prudente, Brazil
| | - Marcos Natal Rufino
- Department of Functional Sciences, Universidade do Oeste Paulista, Presidente Prudente, Brazil
| | - Rogéria Keller
- Department of Functional Sciences, Universidade do Oeste Paulista, Presidente Prudente, Brazil
| | - Hermann Bremer-Neto
- Department of Functional Sciences, Universidade do Oeste Paulista, Faculty of Medicine, Presidente Prudente, Brazil
| |
Collapse
|
35
|
Shirani K, Zanjani BR, Mahmoudi M, Jafarian AH, Hassani FV, Giesy JP, Karimi G. Immunotoxicity of aflatoxin M 1 : as a potent suppressor of innate and acquired immune systems in a subacute study. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2018; 98:5884-5892. [PMID: 30014474 DOI: 10.1002/jsfa.9240] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 05/29/2018] [Accepted: 07/04/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Although, to date, there have been several in vitro and in vivo studies of immunomodulatory effects of aflatoxin M1 (AFB1 ), little is known about the effect of AFM1 on various aspects of innate and acquired immunity. In the present study, AFM1 was administered intraperitoneally, at doses of 25 and 50 μg kg-1 , body mass for 28 days and various immunological parameters were measured. RESULTS Several parameters related to immune function were suppressed: organ mass, cellularity of spleen, proliferation response to lipopolysaccaride and phytohemagglutinin-A, hemagglutination titer, delayed type of hypersensitivity response, spleen cell subtypes, serum hemolytic activity, serum immunoglobulin G level and cytokine production. AFM1 did not cause changes in body mass, hematological parameters or the concentration of immunoglobulin M in blood serum. CONCLUSIONS Overall, the data suggested that AFM1 suppressed innate and acquired immunity. Therefore, with respect to consumer safety, it is extremely important to further control the level of AFM1 in milk, and this should be considered as a precedence for risk management actions. © 2018 Society of Chemical Industry.
Collapse
Affiliation(s)
- Kobra Shirani
- Department of Pharmacodynamics and Toxicology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bamdad R Zanjani
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, School of Medicine, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir H Jafarian
- Cancer Molecular Pathology Research Center, Faculty of Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faezeh V Hassani
- Department of Pharmacodynamics and Toxicology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - John P Giesy
- Department of Veterinary Biomedical Sciences and Toxicology Centre, University of Saskatchewan, Saskatoon, Canada
- Department of Zoology, Center for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- School of Biological Sciences, University of Hong Kong, Hong Kong, China
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
36
|
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: 602] [Impact Index Per Article: 100.3] [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
|