101
|
Okamura T, Hamaguchi M, Nakajima H, Kitagawa N, Majima S, Senmaru T, Okada H, Ushigome E, Nakanishi N, Sasano R, Fukui M. Milk protects against sarcopenic obesity due to increase in the genus Akkermansia in faeces of db/db mice. J Cachexia Sarcopenia Muscle 2023; 14:1395-1409. [PMID: 37132118 PMCID: PMC10235896 DOI: 10.1002/jcsm.13245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/01/2023] [Accepted: 04/03/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Sarcopenic obesity, a combination of sarcopenia and obesity, is a pathological feature of type 2 diabetes. Several human studies have shown that milk is useful in the prevention of sarcopenia. This study was aimed at clarifying the effect of milk on the prevention of sarcopenic obesity in db/db mice. METHODS A randomized and investigator-blinded study was conducted using male db/db mice. Eight-week-old db/db mice were housed for 8 weeks and fed milk (100 μL/day) using a sonde. The faecal microbiota transplantation (FMT) group received antibiotics for 2 weeks, starting at 6 weeks of age, followed by FMT twice a week until 16 weeks of age. RESULTS Milk administration to db/db mice increased grip strength (Milk-: 164.2 ± 4.7 g, Milk+: 230.2 ± 56.0 g, P = 0.017), muscle mass (soleus muscle, Milk-: 164.2 ± 4.7 mg, Milk+: 230.2 ± 56.0 mg, P < 0.001; plantaris muscle, Milk-: 13.3 ± 1.2 mg, Milk+: 16.0 ± 1.7 mg, P < 0.001) and decreased visceral fat mass (Milk-: 2.39 ± 0.08 g, Milk+: 1.98 ± 0.04 mg, P < 0.001), resulting in a significant increase in physical activity (light: P = 0.013, dark: P = 0.034). FMT from mice fed milk not only improved sarcopenic obesity but also significantly improved glucose intolerance. Microarray analysis of gene expression in the small intestine revealed that the expression of amino acid absorption transporter genes, namely, SIc7a5 (P = 0.010), SIc7a1 (P = 0.015), Ppp1r15a (P = 0.041) and SIc7a11 (P = 0.029), was elevated in mice fed milk. In 16S rRNA sequencing of gut microbiota, the genus Akkermansia was increased in both the mice fed milk and the FMT group from the mice fed milk. CONCLUSIONS The findings of this study suggest that besides increasing the intake of nutrients, such as amino acids, milk consumption also changes the intestinal environment, which might contribute to the mechanism of milk-induced improvement of sarcopenic obesity.
Collapse
Affiliation(s)
- Takuro Okamura
- Department of Endocrinology and MetabolismKyoto Prefectural University of Medicine, Graduate School of Medical ScienceKyotoJapan
| | - Masahide Hamaguchi
- Department of Endocrinology and MetabolismKyoto Prefectural University of Medicine, Graduate School of Medical ScienceKyotoJapan
| | - Hanako Nakajima
- Department of Endocrinology and MetabolismKyoto Prefectural University of Medicine, Graduate School of Medical ScienceKyotoJapan
| | - Nobuko Kitagawa
- Department of Endocrinology and MetabolismKyoto Prefectural University of Medicine, Graduate School of Medical ScienceKyotoJapan
| | - Saori Majima
- Department of Endocrinology and MetabolismKyoto Prefectural University of Medicine, Graduate School of Medical ScienceKyotoJapan
| | - Takafumi Senmaru
- Department of Endocrinology and MetabolismKyoto Prefectural University of Medicine, Graduate School of Medical ScienceKyotoJapan
| | - Hiroshi Okada
- Department of Endocrinology and MetabolismKyoto Prefectural University of Medicine, Graduate School of Medical ScienceKyotoJapan
| | - Emi Ushigome
- Department of Endocrinology and MetabolismKyoto Prefectural University of Medicine, Graduate School of Medical ScienceKyotoJapan
| | - Naoko Nakanishi
- Department of Endocrinology and MetabolismKyoto Prefectural University of Medicine, Graduate School of Medical ScienceKyotoJapan
| | | | - Michiaki Fukui
- Department of Endocrinology and MetabolismKyoto Prefectural University of Medicine, Graduate School of Medical ScienceKyotoJapan
| |
Collapse
|
102
|
Zhu X, Shen J, Feng S, Huang C, Wang H, Huo F, Liu H. Akkermansia muciniphila, which is enriched in the gut microbiota by metformin, improves cognitive function in aged mice by reducing the proinflammatory cytokine interleukin-6. MICROBIOME 2023; 11:120. [PMID: 37254162 DOI: 10.1186/s40168-023-01567-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 05/05/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND Metformin, a type 2 diabetes treatment, improves the cognitive function of aged mice; however, whether the protective effects of metformin on cognitive function in aged mice are associated with the gut microbiome is poorly understood. Although some studies suggest that the gut microbe composition influences cognitive function and that manipulating the gut microbiota might protect against age-related cognitive dysfunction, there is no direct evidence to validate that the gut microbiota mediates the effect of metformin on cognitive improvement. RESULTS In this study, we show that the gut microbiota is altered by metformin, which is necessary for protection against ageing-associated cognitive function declines in aged mice. Mice treated with antibiotics did not exhibit metformin-mediated cognitive function protection. Moreover, treatment with Akkermansia muciniphila, which is enriched by metformin, improved cognitive function in aged mice. Mechanistically, A. muciniphila decreased pro-inflammatory-associated pathways, particularly that of the pro-inflammatory cytokine interleukin (IL)-6, in both the peripheral blood and hippocampal profiles, which was correlated with cognitive function improvement. An IL-6 antibody protected cognitive function, and an IL-6 recombinant protein abolished the protective effect of A. muciniphila on cognitive function in aged mice. CONCLUSION This study reveals that A. muciniphila, which is mediated in the gut microbiota by metformin, modulates inflammation-related pathways in the host and improves cognitive function in aged mice by reducing the pro-inflammatory cytokine IL-6. Video Abstract.
Collapse
Affiliation(s)
- Xiaoqi Zhu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Junyan Shen
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Shengyu Feng
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Ce Huang
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Hao Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Fengjiao Huo
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Hailiang Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China.
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, College of Life Sciences, Shihezi University, Shihezi, 832003, China.
- Institute of Advanced Biotechnology, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
103
|
Chen Y, Chen J, Wei H, Gong K, Meng J, Long T, Guo J, Hong J, Yang L, Qiu J, Xiong K, Wang Z, Xu Q. Akkermansia muciniphila-Nlrp3 is involved in the neuroprotection of phosphoglycerate mutase 5 deficiency in traumatic brain injury mice. Front Immunol 2023; 14:1172710. [PMID: 37287985 PMCID: PMC10242175 DOI: 10.3389/fimmu.2023.1172710] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/03/2023] [Indexed: 06/09/2023] Open
Abstract
INTRODUCTION Gut-microbiota-brain axis is a potential treatment to decrease the risk of chronic traumatic encephalopathy following traumatic brain injury (TBI). Phosphoglycerate mutase 5 (PGAM5), a mitochondrial serine/threonine protein phosphatase, resides in mitochondrial membrane and regulates mitochondrial homeostasis and metabolism. Mitochondria mediates intestinal barrier and gut microbiome. OBJECTIVES This study investigated the association between PGAM5 and gut microbiota in mice with TBI. METHODS The controlled cortical impact injury was established in mice with genetically-ablated Pgam5 (Pgam5-/-) or wild type, and WT male mice were treated with fecal microbiota transplantation (FMT) from male Pgam5-/- mice or Akkermansia muciniphila (A. muciniphila). Then the gut microbiota abundance, blood metabolites, neurological function, and nerve injury were detected. RESULTS Treated with antibiotics for suppressing gut microbiota in Pgam5-/- mice partially relieved the role of Pgam5 deficiency in the improvement of initial inflammatory factors and motor dysfunction post-TBI. Pgam5 knockout exhibited an increased abundance of A. muciniphila in mice. FMT from male Pgam5-/- mice enabled better maintenance of amino acid metabolism and peripherial environment than that in TBI-vehicle mice, which suppressed neuroinflammation and improved neurological deficits, and A. muciniphila was negatively associated with intestinal mucosal injury and neuroinflammation post-TBI. Moreover, A. muciniphila treatment ameliorated neuroinflammation and nerve injury by regulating Nlrp3 inflammasome activation in cerebral cortex with TBI. CONCLUSION Thus, the present study provides evidence that Pgam5 is involved in gut microbiota-mediated neuroinflammation and nerve injury, with A. muciniphila-Nlrp3 contributing to peripheral effects.
Collapse
Affiliation(s)
- Yuhua Chen
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou, China
- Department of Central Laboratory, Xi’an Peihua University, Xi’an, Shaanxi, China
- Xiamen Key Laboratory of Brain Center, Department of Neurosurgery, Trauma Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Junhui Chen
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Hong Wei
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou, China
- Department of Rehabilitation Teaching and Research, Xi’an Siyuan University, Xi’an, China
| | - Kai Gong
- Xiamen Key Laboratory of Brain Center, Department of Neurosurgery, Trauma Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Jiao Meng
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou, China
- Department of Central Laboratory, Xi’an Peihua University, Xi’an, Shaanxi, China
| | - Tianlin Long
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou, China
| | - Jianfeng Guo
- Xiamen Key Laboratory of Brain Center, Department of Neurosurgery, Trauma Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Jun Hong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Lingjian Yang
- School of Chemistry & Chemical Engineering, Ankang University, Ankang, China
| | - Junling Qiu
- Department of Cardiology, First Hospital of Northwestern University, Shannxi, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, Hainan, China
- Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, China
| | - Zhanxiang Wang
- Xiamen Key Laboratory of Brain Center, Department of Neurosurgery, Trauma Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Quanhua Xu
- Department of Neurosurgery, Bijie Traditional Chinese Medical Hospital, Bijie, Guizhou, China
| |
Collapse
|
104
|
Deng L, Wojciech L, Png CW, Kioh YQD, Ng GC, Chan ECY, Zhang Y, Gascoigne NRJ, Tan KSW. Colonization with ubiquitous protist Blastocystis ST1 ameliorates DSS-induced colitis and promotes beneficial microbiota and immune outcomes. NPJ Biofilms Microbiomes 2023; 9:22. [PMID: 37185924 PMCID: PMC10130167 DOI: 10.1038/s41522-023-00389-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Blastocystis is a species complex that exhibits extensive genetic diversity, evidenced by its classification into several genetically distinct subtypes (ST). Although several studies have shown the relationships between a specific subtype and gut microbiota, there is no study to show the effect of the ubiquitous Blastocystis ST1 on the gut microbiota and host health. Here, we show that Blastocystis ST1 colonization increased the proportion of beneficial bacteria Alloprevotella and Akkermansia, and induced Th2 and Treg cell responses in normal healthy mice. ST1-colonized mice showed decreases in the severity of DSS-induced colitis when compared to non-colonized mice. Furthermore, mice transplanted with ST1-altered gut microbiota were refractory to dextran sulfate sodium (DSS)-induced colitis via induction of Treg cells and elevated short-chain fat acid (SCFA) production. Our results suggest that colonization with Blastocystis ST1, one of the most common subtypes in humans, exerts beneficial effects on host health through modulating the gut microbiota and adaptive immune responses.
Collapse
Affiliation(s)
- Lei Deng
- Laboratory of Molecular and Cellular Parasitology, Healthy Longevity Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore
| | - Lukasz Wojciech
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore
| | - Chin Wen Png
- Laboratory of Molecular and Cellular Parasitology, Healthy Longevity Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore
| | - Yan Qin Dorinda Kioh
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore
| | - Geok Choo Ng
- Laboratory of Molecular and Cellular Parasitology, Healthy Longevity Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore
| | - Yongliang Zhang
- Laboratory of Molecular and Cellular Parasitology, Healthy Longevity Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore
| | - Nicholas R J Gascoigne
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore
| | - Kevin Shyong Wei Tan
- Laboratory of Molecular and Cellular Parasitology, Healthy Longevity Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore.
| |
Collapse
|
105
|
Shahbazi A, Sepehrinezhad A, Vahdani E, Jamali R, Ghasempour M, Massoudian S, Sahab Negah S, Larsen FS. Gut Dysbiosis and Blood-Brain Barrier Alteration in Hepatic Encephalopathy: From Gut to Brain. Biomedicines 2023; 11:1272. [PMID: 37238943 PMCID: PMC10215854 DOI: 10.3390/biomedicines11051272] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 05/28/2023] Open
Abstract
A common neuropsychiatric complication of advanced liver disease, hepatic encephalopathy (HE), impacts the quality of life and length of hospital stays. There is new evidence that gut microbiota plays a significant role in brain development and cerebral homeostasis. Microbiota metabolites are providing a new avenue of therapeutic options for several neurological-related disorders. For instance, the gut microbiota composition and blood-brain barrier (BBB) integrity are altered in HE in a variety of clinical and experimental studies. Furthermore, probiotics, prebiotics, antibiotics, and fecal microbiota transplantation have been shown to positively affect BBB integrity in disease models that are potentially extendable to HE by targeting gut microbiota. However, the mechanisms that underlie microbiota dysbiosis and its effects on the BBB are still unclear in HE. To this end, the aim of this review was to summarize the clinical and experimental evidence of gut dysbiosis and BBB disruption in HE and a possible mechanism.
Collapse
Affiliation(s)
- Ali Shahbazi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran; (A.S.); (S.M.)
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran;
| | - Ali Sepehrinezhad
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran; (A.S.); (S.M.)
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran;
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad 9919191778, Iran
| | - Edris Vahdani
- Department of Microbiology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 4815733971, Iran;
| | - Raika Jamali
- Research Development Center, Sina Hospital, Tehran University of Medical Sciences, Tehran 1417653761, Iran
- Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran 1417653761, Iran
| | - Monireh Ghasempour
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran;
| | - Shirin Massoudian
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran; (A.S.); (S.M.)
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad 9919191778, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9919191778, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 9815733169, Iran
| | - Fin Stolze Larsen
- Department of Intestinal Failure and Liver Diseases, Rigshospitalet, Inge Lehmanns Vej 5, 2100 Copenhagen, Denmark
| |
Collapse
|
106
|
Guo HH, Shen HR, Wang LL, Luo ZG, Zhang JL, Zhang HJ, Gao TL, Han YX, Jiang JD. Berberine is a potential alternative for metformin with good regulatory effect on lipids in treating metabolic diseases. Biomed Pharmacother 2023; 163:114754. [PMID: 37094549 DOI: 10.1016/j.biopha.2023.114754] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 04/26/2023] Open
Abstract
Metformin (MTF) and berberine (BBR) share several therapeutic benefits in treating metabolic-related disorders. However, as the two agents have very different chemical structure and bioavailability in oral route, the goal of this study is to learn their characteristics in treating metabolic disorders. The therapeutic efficacy of BBR and MTF was systemically investigated in the high fat diet feeding hamsters and/or ApoE(-/-) mice; in parallel, gut microbiota related mechanisms were studied for both agents. We discovered that, although both two drugs had almost identical effects on reducing fatty liver, inflammation and atherosclerosis, BBR appeared to be superior over MTF in alleviating hyperlipidemia and obesity, but MTF was more effective than BBR for the control of blood glucose. Association analysis revealed that the modulation of intestinal microenvironment played a crucial role in the pharmacodynamics of both drugs, in which their respective superiority on the regulation of gut microbiota composition and intestinal bile acids might contribute to their own merits on lowering glucose or lipids. This study shows that BBR may be a good alternative for MTF in treating diabetic patients, especially for those complicated with dyslipidemia and obesity.
Collapse
Affiliation(s)
- Hui-Hui Guo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hao-Ran Shen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lu-Lu Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhi-Gang Luo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jin-Lan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hong-Juan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Tian-Le Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yan-Xing Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Jian-Dong Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
107
|
Zhao J, Yao Y, Li D, Zhu W, Xiao H, Xie M, Xiong Y, Wu J, Ni Q, Zhang M, Xu H. Metagenome and metabolome insights into the energy compensation and exogenous toxin degradation of gut microbiota in high-altitude rhesus macaques (Macaca mulatta). NPJ Biofilms Microbiomes 2023; 9:20. [PMID: 37081021 PMCID: PMC10119431 DOI: 10.1038/s41522-023-00387-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 03/29/2023] [Indexed: 04/22/2023] Open
Abstract
There have been many reports on the genetic mechanism in rhesus macaques (RMs) for environmental adaptation to high altitudes, but the synergistic involvement of gut microbiota in this adaptation remains unclear. Here we performed fecal metagenomic and metabolomic studies on samples from high- and low-altitude populations to assess the synergistic role of gut microbiota in the adaptation of RMs to high-altitude environments. Microbiota taxonomic annotation yielded 7471 microbiota species. There were 37 bacterial species whose abundance was significantly enriched in the high-altitude populations, 16 of which were previously reported to be related to the host's dietary digestion and energy metabolism. Further functional gene enrichment found a stronger potential for gut microbiota to synthesize energy substrate acetyl-CoA using CO2 and energy substrate pyruvate using oxaloacetate, as well as a stronger potential to transform acetyl-CoA to energy substrate acetate in high-altitude populations. Interestingly, there were no apparent differences between low-altitude and high-altitude populations in terms of genes enriched in the main pathways by which the microbiota consumed the three energy substrates, and none of the three energy substrates were detected in the fecal metabolites. These results strongly suggest that gut microbiota plays an important energy compensatory role that helps RMs to adapt to high-altitude environments. Further functional enrichment after metabolite source analysis indicated the abundance of metabolites related to the degradation of exogenous toxins was also significantly higher in high-altitude populations, which suggested a contributory role of gut microbiota to the degradation of exogenous toxins in wild RMs adapted to high-altitude environments.
Collapse
Affiliation(s)
- Junsong Zhao
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
- College of Agronomy and Life Sciences, Zhaotong University, Zhaotong, 657000, China
| | - Yongfang Yao
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Diyan Li
- School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Wei Zhu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Hongtao Xiao
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Meng Xie
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Ying Xiong
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Jiayun Wu
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Qingyong Ni
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Mingwang Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Huailiang Xu
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China.
| |
Collapse
|
108
|
Pellegrino A, Coppola G, Santopaolo F, Gasbarrini A, Ponziani FR. Role of Akkermansia in Human Diseases: From Causation to Therapeutic Properties. Nutrients 2023; 15:nu15081815. [PMID: 37111034 PMCID: PMC10142179 DOI: 10.3390/nu15081815] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The gut microbiota plays a critical role in the modulation of host metabolism and immune response, and its impairment has been implicated in many gastrointestinal and extraintestinal diseases. Current evidence shows the well-documented role of A. muciniphila in maintaining the integrity of the intestinal barrier, modulating the host immune response, and improving several metabolic pathways, making it a key element in the pathogenesis of several human diseases. In this scenario, A. muciniphila is the most promising next-generation probiotic and one of the first microbial species suitable for specific clinical use when compared with traditional probiotics. Further studies are needed to provide more accurate insight into its mechanisms of action and to better elucidate its properties in several major areas, paving the way for a more integrated and personalized therapeutic approach that finally makes the most of our knowledge of the gut microbiota.
Collapse
Affiliation(s)
- Antonio Pellegrino
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino, Gemelli IRCCS, 00168 Rome, Italy
| | - Gaetano Coppola
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino, Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Santopaolo
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino, Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino, Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
109
|
De Filippo C, Costa A, Becagli MV, Monroy MM, Provensi G, Passani MB. Gut microbiota and oleoylethanolamide in the regulation of intestinal homeostasis. Front Endocrinol (Lausanne) 2023; 14:1135157. [PMID: 37091842 PMCID: PMC10113643 DOI: 10.3389/fendo.2023.1135157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/22/2023] [Indexed: 04/08/2023] Open
Abstract
A vast literature strongly suggests that the endocannabinoid (eCB) system and related bioactive lipids (the paracannabinoid system) contribute to numerous physiological processes and are involved in pathological conditions such as obesity, type 2 diabetes, and intestinal inflammation. The gut paracannabinoid system exerts a prominent role in gut physiology as it affects motility, permeability, and inflammatory responses. Another important player in the regulation of host metabolism is the intestinal microbiota, as microorganisms are indispensable to protect the intestine against exogenous pathogens and potentially harmful resident microorganisms. In turn, the composition of the microbiota is regulated by intestinal immune responses. The intestinal microbial community plays a fundamental role in the development of the innate immune system and is essential in shaping adaptive immunity. The active interplay between microbiota and paracannabinoids is beginning to appear as potent regulatory system of the gastrointestinal homeostasis. In this context, oleoylethanolamide (OEA), a key component of the physiological systems involved in the regulation of dietary fat consumption, energy homeostasis, intestinal motility, and a key factor in modulating eating behavior, is a less studied lipid mediator. In the small intestine namely duodenum and jejunum, levels of OEA change according to the nutrient status as they decrease during food deprivation and increase upon refeeding. Recently, we and others showed that OEA treatment in rodents protects against inflammatory events and changes the intestinal microbiota composition. In this review, we briefly define the role of OEA and of the gut microbiota in intestinal homeostasis and recapitulate recent findings suggesting an interplay between OEA and the intestinal microorganisms.
Collapse
Affiliation(s)
- Carlotta De Filippo
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, Pisa, Italy
| | - Alessia Costa
- Dipartimento di Scienze della Salute, Università di Firenze, Firenze, Italy
| | | | - Mariela Mejia Monroy
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche, Pisa, Italy
| | - Gustavo Provensi
- Dipartimento di Neurofarba, Università di Firenze, Firenze, Italy
- *Correspondence: Maria Beatrice Passani, ; Gustavo Provensi,
| | - Maria Beatrice Passani
- Dipartimento di Scienze della Salute, Università di Firenze, Firenze, Italy
- *Correspondence: Maria Beatrice Passani, ; Gustavo Provensi,
| |
Collapse
|
110
|
Xue C, Li G, Gu X, Su Y, Zheng Q, Yuan X, Bao Z, Lu J, Li L. Health and Disease: Akkermansia muciniphila, the Shining Star of the Gut Flora. RESEARCH (WASHINGTON, D.C.) 2023; 6:0107. [PMID: 37040299 PMCID: PMC10079265 DOI: 10.34133/research.0107] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/15/2023] [Indexed: 04/05/2023]
Abstract
Akkermansia muciniphila (A. muciniphila) has drawn much attention as an important gut microbe strain in recent years. A. muciniphila can influence the occurrence and development of diseases of the endocrine, nervous, digestive, musculoskeletal, and respiratory systems and other diseases. It can also improve immunotherapy for some cancers. A. muciniphila is expected to become a new probiotic in addition to Lactobacillus and Bifidobacterium. An increase in A. muciniphila abundance through direct or indirect A. muciniphila supplementation may inhibit or even reverse disease progression. However, some contrary findings are found in type 2 diabetes mellitus and neurodegenerative diseases, where increased A. muciniphila abundance may aggravate the diseases. To enable a more comprehensive understanding of the role of A. muciniphila in diseases, we summarize the relevant information on A. muciniphila in different systemic diseases and introduce regulators of A. muciniphila abundance to promote the clinical transformation of A. muciniphila research.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ganglei Li
- Department of Neurosurgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuanshuai Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiuxian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
111
|
Fan S, Chen S, Lin L. Research progress of gut microbiota and obesity caused by high-fat diet. Front Cell Infect Microbiol 2023; 13:1139800. [PMID: 36992691 PMCID: PMC10040832 DOI: 10.3389/fcimb.2023.1139800] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/28/2023] [Indexed: 03/15/2023] Open
Abstract
Obesity, a chronic metabolic disorder caused by an energy imbalance, has been increasingly prevalent and poses a global health concern. The multifactorial etiology of obesity includes genetics factors, high-fat diet, gut microbiota, and other factors. Among these factors, the implication of gut microbiota in the pathogenesis of obesity has been prominently acknowledged. This study endeavors to investigate the potential contribution of gut microbiota to the development of high-fat diet induced obesity, as well as the current state of probiotic intervention therapy research, in order to provide novel insights for the prevention and management of obesity.
Collapse
Affiliation(s)
- Shuyi Fan
- Scientific Research Department, Brain Hospital of Hunan Province, Second People’s Hospital of Hunan Province, Changsha, Hunan, China
- Department of Clinical Medicine, Xiamen Medical College, Xiamen, Fujian, China
| | - Suyun Chen
- Department of Clinical Medicine, Xiamen Medical College, Xiamen, Fujian, China
| | - Lin Lin
- Scientific Research Department, Brain Hospital of Hunan Province, Second People’s Hospital of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
112
|
Fieschi-Méric L, van Leeuwen P, Denoël M, Lesbarrères D. Encouraging news for in situ conservation: Translocation of salamander larvae has limited impacts on their skin microbiota. Mol Ecol 2023. [PMID: 36872055 DOI: 10.1111/mec.16914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 02/07/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
The key role of symbiotic skin bacteria communities in amphibian resistance to emerging pathogens is well recognized, but factors leading to their dysbiosis are not fully understood. In particular, the potential effects of population translocations on the composition and diversity of hosts' skin microbiota have received little attention, although such transfers are widely carried out as a strategy for amphibian conservation. To characterize the potential reorganization of the microbiota over such a sudden environmental change, we conducted a common-garden experiment simulating reciprocal translocations of yellow-spotted salamander larvae across three lakes. We sequenced skin microbiota samples collected before and 15 days after the transfer. Using a database of antifungal isolates, we identified symbionts with known function against the pathogen Batrachochytrium dendrobatidis, a major driver of amphibian declines. Our results indicate an important reorganization of bacterial assemblages throughout ontogeny, with strong changes in composition, diversity and structure of the skin microbiota in both control and translocated individuals over the 15 days of monitoring. Unexpectedly, the diversity and community structure of the microbiota were not significantly affected by the translocation event, thus suggesting a strong resilience of skin bacterial communities to environmental change-at least across the time-window studied here. A few phylotypes were more abundant in the microbiota of translocated larvae, but no differences were found among pathogen-inhibiting symbionts. Taken together, our results support amphibian translocations as a promising strategy for this endangered animal class, with limited impact on their skin microbiota.
Collapse
Affiliation(s)
- Léa Fieschi-Méric
- Laboratory of Ecology and Conservation of Amphibians (LECA), Freshwater and OCeanic science Unit of reSearch (FOCUS), Université de Liège, Liège, Belgium.,Biology Department, Laurentian University, Sudbury, Ontario, Canada
| | - Pauline van Leeuwen
- Biology Department, Laurentian University, Sudbury, Ontario, Canada.,Conservation Genetics Laboratory, University de Liège, Liège, Belgium
| | - Mathieu Denoël
- Laboratory of Ecology and Conservation of Amphibians (LECA), Freshwater and OCeanic science Unit of reSearch (FOCUS), Université de Liège, Liège, Belgium
| | - David Lesbarrères
- Biology Department, Laurentian University, Sudbury, Ontario, Canada.,Environment and Climate Change Canada, Ottawa, Ontario, Canada
| |
Collapse
|
113
|
Xu R, Zhang Y, Chen S, Zeng Y, Fu X, Chen T, Luo S, Zhang X. The role of the probiotic Akkermansia muciniphila in brain functions: insights underpinning therapeutic potential. Crit Rev Microbiol 2023; 49:151-176. [PMID: 35272549 DOI: 10.1080/1040841x.2022.2044286] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The role of Akkermansia muciniphila, one of the most abundant microorganisms of the intestinal microbiota, has been studied extensively in metabolic diseases, such as obesity and diabetes. It is considered a next-generation probiotic microorganism. Although its mechanism of action has not been fully elucidated, accumulating evidence indicates the important role of A. muciniphila in brain functions via the gut-brain axis and its potential as a therapeutic target in various neuropsychiatric disorders. However, only a limited number of studies, particularly clinical studies, have directly assessed the therapeutic effects of A. muciniphila interventions in these disorders. This is the first review to discuss the comprehensive mechanism of A. muciniphila in the gut-brain axis via the protection of the intestinal mucosal barrier and modulation of the immune system and metabolites, such as short-chain fatty acids, amino acids, and amino acid derivatives. Additionally, the role of A. muciniphila and its therapeutic potential in various neuropsychiatric disorders, including Alzheimer's disease and cognitive deficit, amyotrophic lateral sclerosis, Parkinson's disease, and multiple sclerosis, have been discussed. The review suggests the potential role of A. muciniphila in healthy brain functions.
Collapse
Affiliation(s)
- Ruiling Xu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinic Research Center for Mental Disorders, Changsha, Hunan, China.,National Technology Institute on Mental Disorders, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China.,Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yuxuan Zhang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinic Research Center for Mental Disorders, Changsha, Hunan, China.,National Technology Institute on Mental Disorders, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China.,Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha, China
| | - Shurui Chen
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinic Research Center for Mental Disorders, Changsha, Hunan, China.,National Technology Institute on Mental Disorders, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China.,Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yaohui Zeng
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinic Research Center for Mental Disorders, Changsha, Hunan, China.,National Technology Institute on Mental Disorders, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China.,Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xuan Fu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinic Research Center for Mental Disorders, Changsha, Hunan, China.,National Technology Institute on Mental Disorders, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China.,Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ti Chen
- Clinical Laboratory, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shilin Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaojie Zhang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinic Research Center for Mental Disorders, Changsha, Hunan, China.,National Technology Institute on Mental Disorders, Changsha, Hunan, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan, China.,Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
114
|
Altered gastrointestinal tract structure and microbiome following cerebral malaria infection. Parasitol Res 2023; 122:789-799. [PMID: 36602586 DOI: 10.1007/s00436-022-07775-2] [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: 06/07/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023]
Abstract
Cerebral malaria (CM) is the most severe form of malaria with the highest mortality rate and can result in life-long neurological deficits and ongoing comorbidities. Factors contributing to severity of infection and development of CM are not fully elucidated. Recent studies have indicated a key role of the gut microbiome in a range of health conditions that affect the brain, but limited microbiome research has been conducted in the context of malaria. To address this knowledge gap, the impact of CM on the gut microbiome was investigated in mice. C57BL/6J mice were infected with Plasmodium berghei ANKA (PbA) parasites and compared to non-infected controls. Microbial DNA from faecal pellets collected daily for 6-days post-infection were extracted, and microbiome comparisons conducted using 16S rRNA profiling. We identified significant differences in the composition of bacterial communities between the infected and the non-infected groups, including a higher abundance of the genera Akkermansia, Alistipes and Alloprevotella in PbA-infected mice. Furthermore, intestinal samples were collected post-cull for morphological analysis. We determined that the caecal weight was significantly lower, and the small intestine was significantly longer in PbA-infected mice than in the non-infected controls. We concluded that changes in microbial community composition were primarily driven by the infection protocol and, to a lesser extent, by the time of infection. Our findings pave the way for a new area of research and novel intervention strategies to modulate the severity of cerebral malaria disease.
Collapse
|
115
|
Peng M, Yi W, Murong M, Peng N, Tong H, Jiang M, Jin D, Peng S, Liang W, Quan J, Li M, Shi L, Xiao G. Akkermansia muciniphila improves heat stress-impaired intestinal barrier function by modulating HSP27 in Caco-2 cells. Microb Pathog 2023; 177:106028. [PMID: 36796737 DOI: 10.1016/j.micpath.2023.106028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/18/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023]
Abstract
OBJECTIVE Heat stress causes an elevation of intestinal epithelial barrier permeability and leads to multiple organ dysfunction in heatstroke. Akkermansia muciniphila (A. muciniphila) plays a role in maintaining intestinal integrity and improving the inflammatory state. This study aimed to investigate whether A. muciniphila could alleviate heat stress-induced dysfunction of intestinal permeability in Caco-2 monolayers and have the preventive effects on heatstroke. METHODS Human intestinal epithelial Caco-2 cells were preincubated with live or pasteurized A. muciniphila then exposed to heat stress at 43 °C. Transepithelial electrical resistance (TEER) and the flux of horseradish peroxidase (HRP) across cell monolayers were measured to determine intestinal permeability. The levels of the tight junction proteins Occludin, ZO-1 and HSP27 were analyzed by Western blotting. These proteins were immunostained and localized by fluorescence microscopy. TJ morphology was observed using transmission electron microscopy (TEM). RESULTS Both live and pasteurized A. muciniphila effectively attenuated the decrease in TEER and impairment of intestinal permeability in HRP flux induced by heat exposure. A. muciniphila significantly elevated the expression of Occludin and ZO-1 by promoting HSP27 phosphorylation. The distortion and redistribution of tight junction proteins and disruption of morphology were also effectively prevented by pretreatment with A. muciniphila. CONCLUSION This study indicates for the first time that both live and pasteurized A. muciniphila play an important protective role against heat-induced permeability dysfunction and epithelial barrier damage.
Collapse
Affiliation(s)
- Mian Peng
- Department of Critical Care Medicine, Shenzhen Luohu Hospital Group, Shenzhen, Guangdong, 518001, China
| | - Wanhua Yi
- Department of Critical Care Medicine, General Hospital of Southern Theater Command, Guangzhou, Guangdong, 510010, China
| | - Min Murong
- Department of Nutrition, General Hospital of Southern Theater Command, Guangzhou, Guangdong, 510010, China
| | - Na Peng
- Emergency Department, General Hospital of Southern Theater Command, Guangzhou, Guangdong, 510010, China
| | - Huasheng Tong
- Emergency Department, General Hospital of Southern Theater Command, Guangzhou, Guangdong, 510010, China
| | - Mengliu Jiang
- Department of Nutrition, General Hospital of Southern Theater Command, Guangzhou, Guangdong, 510010, China
| | - Di Jin
- Department of Nutrition, General Hospital of Southern Theater Command, Guangzhou, Guangdong, 510010, China
| | - Suliu Peng
- Department of Nutrition, General Hospital of Southern Theater Command, Guangzhou, Guangdong, 510010, China
| | - Weifen Liang
- Department of Nutrition, General Hospital of Southern Theater Command, Guangzhou, Guangdong, 510010, China
| | - Jingwen Quan
- Department of Nutrition, General Hospital of Southern Theater Command, Guangzhou, Guangdong, 510010, China
| | - Muman Li
- Department of Nutrition, General Hospital of Southern Theater Command, Guangzhou, Guangdong, 510010, China
| | - Linna Shi
- Department of Nutrition, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Guizhen Xiao
- Department of Nutrition, General Hospital of Southern Theater Command, Guangzhou, Guangdong, 510010, China; Huabo Post-Doctoral Research Center, Biological Pharmaceutical Research Institute, Guangzhou, Guangdong, 510010, China.
| |
Collapse
|
116
|
Zhao Q, Yu J, Hao Y, Zhou H, Hu Y, Zhang C, Zheng H, Wang X, Zeng F, Hu J, Gu L, Wang Z, Zhao F, Yue C, Zhou P, Zhang H, Huang N, Wu W, Zhou Y, Li J. Akkermansia muciniphila plays critical roles in host health. Crit Rev Microbiol 2023; 49:82-100. [PMID: 35603929 DOI: 10.1080/1040841x.2022.2037506] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Akkermansia muciniphila, an intestinal microorganism, belongs to Verrucomicrobia, one of the most abundant microorganisms in the mammalian gut. It is a mucin-degrading bacterium that can colonise intestines of mammals such as humans and mice by utilising mucin as the only nitrogen and carbon source. When A. muciniphila colonises the intestine, its metabolites interact with the intestinal barrier, affecting host health by consolidating the intestinal barrier, regulating metabolic functions of the intestinal and circulatory systems, and regulating immune functions. This review summarised the mechanisms of A. muciniphila-host interactions that are relevant to host health. We focussed on characteristics of A. muciniphila in relation to its metabolites to provide a comprehensive understanding of A. muciniphila and its effects on host health and disease processes.
Collapse
Affiliation(s)
- Qixiang Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jiadong Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yan Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Hong Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yawen Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Chen Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huaping Zheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xiaoyan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Fanlian Zeng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jing Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Linna Gu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Zhen Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Fulei Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Chengcheng Yue
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Pei Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Haozhou Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Nongyu Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wenling Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yifan Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jiong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
117
|
Sciascia S, Arbrile M, Trunfio M, Calcagno A, Radin M, Roccatello D, Lembo D, Civra A. The role of bacteria and viruses in Behçet syndrome: Should we move towards new paradigms? Autoimmun Rev 2023; 22:103237. [PMID: 36414220 DOI: 10.1016/j.autrev.2022.103237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/17/2022] [Indexed: 11/21/2022]
Abstract
The etiological complexity of Behçet syndrome (BS), an immune-mediated rare form of vasculitis characterized by multi-organ involvement, is still elusive due to an incomplete understanding of the synergy between genetic susceptibility, environmental triggers, and an abnormal immune response. Long-standing theories regarding the origins of BS include the involvement of infectious organisms supporting an aberrant immunological response through different mechanisms, including molecular mimicry. Additionally, it has been demonstrated that the BS phenotypes are linked to oral and gut microbiome dysbiosis, which is a dynamic reservoir of millions of microbes containing proteins and metabolites that can mimic the autoantigens. Infections, including viral pathogens, could potentially trigger the inflammation and symptoms of BS. In this review, we aim to describe the available evidence on the cross-talk between BS and infections in order to discuss potential clinical implications and possible therapeutic targets.
Collapse
Affiliation(s)
- Savino Sciascia
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley (North-West Italy), San Giovanni Bosco Hub Hospital, Department of Clinical and Biological Sciences of the University of Turin, Turin, Italy.
| | - Marta Arbrile
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley (North-West Italy), San Giovanni Bosco Hub Hospital, Department of Clinical and Biological Sciences of the University of Turin, Turin, Italy
| | - Mattia Trunfio
- Unit of Infectious diseases, Department of medical sciences at Amedeo di Savoia Hospital, University of Turin, Turin, Italy
| | - Andrea Calcagno
- Unit of Infectious diseases, Department of medical sciences at Amedeo di Savoia Hospital, University of Turin, Turin, Italy
| | - Massimo Radin
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley (North-West Italy), San Giovanni Bosco Hub Hospital, Department of Clinical and Biological Sciences of the University of Turin, Turin, Italy
| | - Dario Roccatello
- University Center of Excellence on Nephrologic, Rheumatologic and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) with Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley (North-West Italy), San Giovanni Bosco Hub Hospital, Department of Clinical and Biological Sciences of the University of Turin, Turin, Italy
| | - David Lembo
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Andrea Civra
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
118
|
Xie R, Zhang H, Zhang H, Li C, Cui D, Li S, Li Z, Liu H, Huang J. Hemagglutinin expressed by yeast reshapes immune microenvironment and gut microbiota to trigger diverse anti-infection response in infected birds. Front Immunol 2023; 14:1125190. [PMID: 37143654 PMCID: PMC10151582 DOI: 10.3389/fimmu.2023.1125190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/22/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction The H5N8 influenza virus is a highly pathogenic pathogen for poultry and human. Vaccination is the most effective method to control the spread of the virus right now. The traditional inactivated vaccine, though well developed and used widely, is laborious during application and more interests are stimulated in developing alternative approaches. Methods In this study, we developed three hemagglutinin (HA) gene-based yeast vaccine. In order to explore the protective efficacy of the vaccines, the gene expression level in the bursa of Fabricius and the structure of intestinal microflora in immunized animals were analyzed by RNA seq and 16SrRNA sequencing, and the regulatory mechanism of yeast vaccine was evaluated. Results All of these vaccines elicited the humoral immunity, inhibited viral load in the chicken tissues, and provided partial protective efficacy due to the high dose of the H5N8 virus. Molecular mechanism studies suggested that, compared to the traditional inactivated vaccine, our engineered yeast vaccine reshaped the immune cell microenvironment in bursa of Fabricius to promote the defense and immune responses. Analysis of gut microbiota further suggested that oral administration of engineered ST1814G/H5HA yeast vaccine increased the diversity of gut microbiota and the increasement of Reuteri and Muciniphila might benefit the recovery from influenza virus infection. These results provide strong evidence for further clinical use of these engineered yeast vaccine in poultry.
Collapse
Affiliation(s)
- Ruyu Xie
- School of Life Science, Tianjin University, Tianjin, China
| | - Huixia Zhang
- School of Life Science, Tianjin University, Tianjin, China
| | - Han Zhang
- School of Life Science, Tianjin University, Tianjin, China
| | - Changyan Li
- School of Life Science, Tianjin University, Tianjin, China
| | - Daqing Cui
- School of Life Science, Tianjin University, Tianjin, China
| | - Shujun Li
- School of Life Science, Tianjin University, Tianjin, China
| | - Zexing Li
- School of Life Science, Tianjin University, Tianjin, China
| | - Hualei Liu
- China Animal Health and Epidemiology Center, Qingdao, Shandong, China
- *Correspondence: Hualei Liu, ; Jinhai Huang,
| | - Jinhai Huang
- School of Life Science, Tianjin University, Tianjin, China
- *Correspondence: Hualei Liu, ; Jinhai Huang,
| |
Collapse
|
119
|
Zhao L, Qiu Y, Zhang P, Wu X, Zhao Z, Deng X, Yang L, Wang D, Yuan G. Gut microbiota mediates positive effects of liraglutide on dyslipidemia in mice fed a high-fat diet. Front Nutr 2022; 9:1048693. [PMID: 36643973 PMCID: PMC9835552 DOI: 10.3389/fnut.2022.1048693] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/14/2022] [Indexed: 12/30/2022] Open
Abstract
Except for improving glycemic control, liraglutide, one of the glucagon-like peptide-1 receptor agonists, has exerted promising therapeutic effects for dyslipidemia. It has been proved that gut microbiota plays a dramatic role in regulating lipid metabolism. This study aims to explore whether liraglutide could improve dyslipidemia by modulating the gut microbiota in mice fed a high-fat diet (HFD). The C57BL/6 mice were fed a HFD to establish an animal model of dyslipidemia, and then administered with liraglutide or normal saline (NS) for 12 weeks. Indices of glucolipid metabolism were evaluated. Gut microbiota of the mice was analyzed by 16S rRNA gene sequencing. Compared with HFD group, liraglutide significantly alleviated weight, total cholesterol (TC) and low-density lipoprotein cholesterol (LDL) levels, meanwhile elevating high-density lipoprotein cholesterol (HDL) levels (all p < 0.05). The gut microbiota analysis revealed that liraglutide greatly reduced the relative abundance of Firmicutes and augmented that of Bacteroidetes, with a concomitant drop in the Firmicutes/Bacteroidetes ratio. Meanwhile, liraglutide dramatically changed the overall composition, promoted the growth of beneficial microbes (Akkermansia, Lactobacillus, Parabacteroides, Oscillospira, etc.), and inhibited the growth of harmful microbes (AF12, Shigella, Proteobacteria, Xenorhabdus, etc.). Especially, the relative abundance of Akkermansia increased the most after liraglutide treatment. Correlation analysis suggested that TC and LDL were positively correlated with some harmful bacteria, and negatively associated with beneficial bacteria. This study confirmed that liraglutide had a certain therapeutic effect on dyslipidemia in HFD-fed mice and could regulate the composition of the gut microbiota associated with lipid metabolism, especially Akkermansia. Thus, affecting gut microbiota might be a potential mechanism of liraglutide in attenuating dyslipidemia.
Collapse
Affiliation(s)
- Li Zhao
- Department of Endocrinology and Metabolism, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China,*Correspondence: Li Zhao,
| | - Yue Qiu
- Department of Endocrinology and Metabolism, The First People’s Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Panpan Zhang
- Department of Endocrinology, Taicang Hospital of Traditional Chinese Medicine, Taicang, Jiangsu, China
| | - Xunan Wu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhicong Zhao
- Department of Endocrinology and Metabolism, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xia Deng
- Department of Endocrinology and Metabolism, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ling Yang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Dong Wang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guoyue Yuan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China,Guoyue Yuan,
| |
Collapse
|
120
|
Metabolic Fate of Orally Ingested Proanthocyanidins through the Digestive Tract. Antioxidants (Basel) 2022; 12:antiox12010017. [PMID: 36670878 PMCID: PMC9854439 DOI: 10.3390/antiox12010017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Proanthocyanidins (PACs), which are oligomers or polymers of flavan-3ols with potent antioxidative activity, are well known to exert a variety of beneficial health effects. Nonetheless, their bioaccessibility and bioavailability have been poorly assessed. In this review, we focused on the metabolic fate of PACs through the digestive tract. When oligomeric and polymeric PACs are orally ingested, a large portion of the PACs reach the colon, where a small portion is subjected to microbial degradation to phenolic acids and valerolactones, despite the possibility that slight depolymerization of PACs occurs in the stomach and small intestine. Valerolactones, as microbiota-generated catabolites of PACs, may contribute to some of the health benefits of orally ingested PACs. The remaining portion interacts with gut microbiota, resulting in improved microbial diversity and, thereby, contributing to improved health. For instance, an increased amount of beneficial gut bacteria (e.g., Akkermansia muciniphila and butyrate-producing bacteria) could ameliorate host metabolic functions, and a lowered ratio of Firmicutes/Bacteroidetes at the phylum level could mitigate obesity-related metabolic disorders.
Collapse
|
121
|
Reclassification of eight Akkermansia muciniphila strains and description of Akkermansia massiliensis sp. nov. and Candidatus Akkermansia timonensis, isolated from human feces. Sci Rep 2022; 12:21747. [PMID: 36526682 PMCID: PMC9758162 DOI: 10.1038/s41598-022-25873-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Akkermansia muciniphila is a human intestinal tract bacterium that plays an important role in the mucus layer renewal. Several studies have demonstrated that it is a modulator for gut homeostasis and a probiotic for human health. The Akkermansia genus contains two species with standing in nomenclature but their genomic diversity remains unclear. In this study, eight new Akkermansia sp. strains were isolated from the human gut. Using the digital DNA-DNA hybridization (dDDH), average nucleotide identity (ANI) and core genome-based phylogenetic analysis applied to 104 A. muciniphila whole genomes sequences, strains were reclassified into three clusters. Cluster I groups A. muciniphila strains (including strain ATCC BAA-835T as type strain), whereas clusters II and III represent two new species. A member of cluster II, strain Marseille-P6666 differed from A. muciniphila strain ATCC BAA-835T and from A. glycaniphila strain PytT in its ability to grow in microaerophilic atmosphere up to 42 °C, to assimilate various carbon sources and to produce acids from a several compounds. The major fatty acids of strain Marseille-P6666 were 12-methyl-tetradecanoic and pentadecanoic acids. The DNA G + C content of strain Marseille-P6666 was 57.8%. On the basis of these properties, we propose the name A. massiliensis sp. nov. for members of cluster II, with strain Marseille-P6666T (= CSUR P6666 = CECT 30548) as type strain. We also propose the name "Candidatus Akkermansia timonensis" sp. nov. for the members of cluster III, which contains only uncultivated strains, strain Akk0196 being the type strain.
Collapse
|
122
|
Bertoncini-Silva C, Zingg JM, Fassini PG, Suen VMM. Bioactive dietary components-Anti-obesity effects related to energy metabolism and inflammation. Biofactors 2022; 49:297-321. [PMID: 36468445 DOI: 10.1002/biof.1921] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/18/2022] [Indexed: 12/10/2022]
Abstract
Obesity is the result of the long-term energy imbalance between the excess calories consumed and the few calories expended. Reducing the intake of energy dense foods (fats, sugars), and strategies such as fasting and caloric restriction can promote body weight loss. Not only energy in terms of calories, but also the specific composition of the diet can affect the way the food is absorbed and how its energy is stored, used or dissipated. Recent research has shown that bioactive components of food, such as polyphenols and vitamins, can influence obesity and its pathologic complications such as insulin resistance, inflammation and metabolic syndrome. Individual micronutrients can influence lipid turnover but for long-term effects on weight stability, dietary patterns containing several micronutrients may be required. At the molecular level, these molecules modulate signaling and the expression of genes that are involved in the regulation of energy intake, lipid metabolism, adipogenesis into white, beige and brown adipose tissue, thermogenesis, lipotoxicity, adipo/cytokine synthesis, and inflammation. Higher concentrations of these molecules can be reached in the intestine, where they can modulate the composition and action of the microbiome. In this review, the molecular mechanisms by which bioactive compounds and vitamins modulate energy metabolism, inflammation and obesity are discussed.
Collapse
Affiliation(s)
- Caroline Bertoncini-Silva
- Department of Internal Medicine, Division of Nutrology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Jean-Marc Zingg
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Priscila Giacomo Fassini
- Department of Internal Medicine, Division of Nutrology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Vivian Marques Miguel Suen
- Department of Internal Medicine, Division of Nutrology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
123
|
Chen L, Liu Y, Tang Z, Song Z, Cao F, Shi X, Xie P, Wei P, Li M. Radix Angelica dahuricae extract ameliorates oestrogen deficiency-induced dyslipidaemia in ovariectomized (OVX) rats by modulating the gut microbiota and bile acid signalling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154440. [PMID: 36162241 DOI: 10.1016/j.phymed.2022.154440] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 06/23/2022] [Accepted: 09/04/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Radix Angelica dahuricae (RAD), a well-known traditional Chinese medicine, displays a promising effect on alleviating lipid metabolism. However, the improvement of RAD on oestrogen deficiency-induced dyslipidaemia and the underlying mechanism are unclear. PURPOSE The aim of this study was to study the effect of RAD on oestrogen deficiency-induced dyslipidaemia in ovariectomized (OVX) rats and investigate the involvement of the gut microbiota and bile acid signalling in the protective effects. METHODS Bilateral ovariectomy was executed to establish an oestrogen deficiency model. Serum biochemical indexes, liver lipids, inflammatory cytokines and histomorphology were evaluated. Gut microbes were analysed via 16S rRNA sequencing. Faecal short-chain fatty acids (SCFAs) and serum bile acids were quantified by gas chromatography-flame ionization detection (GC-FID) and ultra-high-performance chromatography-tandem mass spectrometry (UPLC-MS/MS), respectively. The expression of genes related to bile acid synthesis, metabolism and enterohepatic circulation in the liver and caecum was measured by real-time PCR. RESULTS The results displayed that RAD administration markedly decreased body weight, TC and TG levels in the serum and liver, and hepatic steatosis and inflammation in OVX rats. RAD administration could significantly regulate the gut microbial composition, increasing the abundance of Lactobacillus, increasing the content of bile salt hydrolase (BSH), and reestablishing the SCFA profile and bile acid metabolism profile in OVX rats. RAD administration could increase the gene expression of HMG-CoA reductase (HMGCR) and cytochrome P450 7A1(CYP7A1) and regulate the gene expression of the related receptors as well as proteins in enterohepatic circulation. CONCLUSIONS RAD alleviated oestrogen deficiency-induced dyslipidaemia in OVX rats. Modulation of the gut microbiota composition and bile acid signalling may be the underlying mechanism.
Collapse
Affiliation(s)
- Lin Chen
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, P R China.
| | - Yanru Liu
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, P R China.
| | - Zhishu Tang
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, P R China; China Academy of Chinese Medical Sciences, Beijing 100700, P R China.
| | - Zhongxing Song
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, P R China
| | - Fan Cao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, P R China
| | - Xinbo Shi
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, P R China
| | - Pei Xie
- Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi & Education Ministry, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Innovative Drug Research Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712083, P R China
| | - Peifeng Wei
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, P R China
| | - Min Li
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi 712046, P R China
| |
Collapse
|
124
|
Tursi A, Papa V, Lopetuso LR, Settanni CR, Gasbarrini A, Papa A. Microbiota Composition in Diverticular Disease: Implications for Therapy. Int J Mol Sci 2022; 23:14799. [PMID: 36499127 PMCID: PMC9736941 DOI: 10.3390/ijms232314799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
Gut microbiota (GM) composition and its imbalance are crucial in the pathogenesis of several diseases, mainly those affecting the gastrointestinal tract. Colon diverticulosis and its clinical manifestations (diverticular disease, DD) are among the most common digestive disorders in developed countries. In recent literature, the role of GM imbalance in the onset of the different manifestations within the clinical spectrum of DD has been highlighted. This narrative review aims to summarize and critically analyze the current knowledge on GM dysbiosis in diverticulosis and DD by comparing the available data with those found in inflammatory bowel disease (IBD). The rationale for using probiotics to rebalance dysbiosis in DD is also discussed.
Collapse
Affiliation(s)
- Antonio Tursi
- Territorial Gastroenterology Service, ASL BAT, 70031 Andria, Italy
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University, 00168 Rome, Italy
| | - Valerio Papa
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University, 00168 Rome, Italy
- Digestive Surgery, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
| | - Loris Riccardo Lopetuso
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Gemelli, IRCCS, 00168 Rome, Italy
- Department of Medicine and Ageing Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Carlo Romano Settanni
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Gemelli, IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University, 00168 Rome, Italy
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Gemelli, IRCCS, 00168 Rome, Italy
| | - Alfredo Papa
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University, 00168 Rome, Italy
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Gemelli, IRCCS, 00168 Rome, Italy
| |
Collapse
|
125
|
Deledda A, Palmas V, Heidrich V, Fosci M, Lombardo M, Cambarau G, Lai A, Melis M, Loi E, Loviselli A, Manzin A, Velluzzi F. Dynamics of Gut Microbiota and Clinical Variables after Ketogenic and Mediterranean Diets in Drug-Naïve Patients with Type 2 Diabetes Mellitus and Obesity. Metabolites 2022; 12:1092. [PMID: 36355175 PMCID: PMC9693465 DOI: 10.3390/metabo12111092] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/29/2022] [Accepted: 11/08/2022] [Indexed: 07/30/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM), the most common form of diabetes, is a progressive chronic metabolic disease that has increasingly spread worldwide, enhancing the mortality rate, particularly from cardiovascular diseases (CVD). Lifestyle improvement through diet and physical activity is, together with drug treatment, the cornerstone of T2DM management. The Mediterranean diet (MD), which favors a prevalence of unprocessed vegetable foods and a reduction in red meats and industrial foods, without excluding any food category, is usually recommended. Recently, scientific societies have promoted a very low-calorie ketogenic diet (VLCKD), a multiphasic protocol that limits carbohydrates and then gradually re-introduces them, with a favorable outcome on body weight and metabolic parameters. Indeed, gut microbiota (GM) modifications have been linked to overweight/obesity and metabolic alterations typical of T2DM. Diet is known to affect GM largely, but only a few studies have investigated the effects of VLCKD on GM, especially in T2DM. In this study, we have compared anthropometric, biochemical, lifestyle parameters, the quality of life, and the GM of eleven patients with recently diagnosed T2DM and overweight or obesity, randomly assigned to two groups of six and five patients who followed the VLCKD (KETO) or hypocaloric MD (MEDI) respectively; parameters were recorded at baseline (T0) and after two (T2) and three months (T3). The results showed that VLCKD had more significant beneficial effects than MD on anthropometric parameters, while biochemical improvements did not statistically differ. As for the GM, despite the lack of significant results regarding the alpha and beta diversity, and the Firmicutes/Bacteroidota ratio between the two groups, in the KETO group, a significant increase in beneficial microbial taxa such as Verrucomicrobiota phylum with its members Verrucomicrobiae, Verrucomicrobiales, Akkermansiaceae, and Akkermansia, Christensenellaceae family, Eubacterium spp., and a reduction in microbial taxa previously associated with obesity (Firmicutes and Actinobacteriota) or other diseases (Alistipes) was observed both at T2 and T3. With regards to the MEDI group, variations were limited to a significant increase in Actinobacteroidota phylum at T2 and T3 and Firmicutes phylum at T3. Moreover, a metagenomic alteration linked to some metabolic pathways was found exclusively in the KETO group. In conclusion, both dietary approaches allowed patients to improve their state of health, but VLCKD has shown better results on body composition as well as on GM profile.
Collapse
Affiliation(s)
- Andrea Deledda
- Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Vanessa Palmas
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Vitor Heidrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo 05508-900, Brazil
- Centro de Oncologia Molecular, Hospital Sírio-Libanês, São Paulo 01308-050, Brazil
| | - Michele Fosci
- Endocrinology Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy
| | - Mauro Lombardo
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Giulia Cambarau
- Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Alessio Lai
- Diabetologia, P.O. Binaghi, ASSL Cagliari, 09126 Cagliari, Italy
| | - Marietta Melis
- Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Elisabetta Loi
- Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Andrea Loviselli
- Endocrinology Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy
| | - Aldo Manzin
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Fernanda Velluzzi
- Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| |
Collapse
|
126
|
Li Z, Ke H, Wang Y, Chen S, Liu X, Lin Q, Wang P, Chen Y. Global trends in Akkermansia muciniphila research: A bibliometric visualization. Front Microbiol 2022; 13:1037708. [PMID: 36439840 PMCID: PMC9685322 DOI: 10.3389/fmicb.2022.1037708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/28/2022] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Akkermansia muciniphila is a member of the gut microbiome, using mucin as sources of carbon, nitrogen, and energy. Since the first discovery of this unique bacterium in 2004, A. muciniphila has been extensively studied. It is considered a promising "next-generation beneficial microbe." The purpose of this paper is to sort out the research status and summarize the hotspots through bibliometric analysis of the publications of A. muciniphila. METHODS The publications about A. muciniphila from January 2004 to February 2022 were obtained from the Web of Science Core Collection. Visualization analyses were performed using three bibliometric tools and GraphPad Prism. RESULTS A total of 1,478 published documents were analyzed. Annual publication number grew from 1 in 2004 to 336 in 2021, with China being the leading producer (33.36%). De Vos, Willem M was the most productive author with the highest H-index (documents = 56, H-index = 37), followed by Cani, Patrice D (documents = 35, H-index = 25). And Scientific Reports published the most papers. PNAS was the keystone taxa in this field, with high betweenness centrality (0.11) and high frequency. The keywords with high frequency in recent years include: oxidative stress, diet, metformin, fecal microbiota transplantation, short-chain fatty acids, polyphenols, microbiota metabolites and so on. The keyword "oxidative stress" was observed to be increasing in frequency recently. CONCLUSION Over time, the scope of the research on the clinical uses of A. muciniphila has gradually increased, and was gradually deepened and developed toward a more precise level. A. muciniphila is likely to remain a research hotspot in the foreseeable future and may contribute to human health.
Collapse
Affiliation(s)
- Zitong Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haoran Ke
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuze Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiuying Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianyun Lin
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Pu Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Integrative Microecology Center, Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
127
|
Wang B, Wu C, Cui L, Wang H, Liu Y, Cui W. Dietary aluminium intake disrupts the overall structure of gut microbiota in Wistar rats. Food Sci Nutr 2022; 10:3574-3584. [PMID: 36348807 PMCID: PMC9632190 DOI: 10.1002/fsn3.2955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/18/2022] [Accepted: 05/26/2022] [Indexed: 12/05/2022] Open
Abstract
Approximately, 40% of ingested dietary aluminium accumulates in the intestine, which has been considered a target organ for dietary aluminium exposure. The gut microbiota may be the first protective barrier against the toxic metal aluminium and a crucial mediator of the bioavailability of metal aluminium. We previously evaluated dietary aluminium intake and its health risks in a population from Jilin Province, China, and found that the average daily intake of aluminium in the diet of residents in Jilin Province was 0.163 mg/kg after the total diet survey. In the present study, the equivalent concentration of aluminium in rats was extrapolated by the average dietary aluminium intake in the population of Jilin Province based on body surface area. Furthermore, healthy adult Wistar rats were randomly divided into four groups (n = 15 for each group): a control group and three groups treated with aluminium solution (1, 10, and 100 mg/kg/day, intragastrically) for 28 days. Following treatment, necrosis of renal tubular epithelial cells, hyperplasia of bile ducts and hyperplasia of heart tissue, as well as fiber in the liver, kidney, and heart tissues of aluminium-treated rats were observed, although there were no significant changes in the spleen and brain. Subsequently, fecal samples were withdrawn for 16S rRNA gene sequence analysis. It was found that aluminium decreased the microbiota diversity and changed the overall community structure of the gut microbiota, including three phyla and four genera, together with the regulation of 12 signaling pathways. Collectively, treatment with aluminium markedly altered the structure of the gut microbiota, suggesting that the disorders of intestinal flora induced by aluminium may be an important mechanism for aluminium toxicity.
Collapse
Affiliation(s)
- Bo Wang
- Department of Nutrition and Food Hygiene, School of Public HealthJilin UniversityChangchunChina
- Department of Physical and Chemical TestJilin Provincial Center for Disease Control and PreventionChangchunChina
| | - Caihong Wu
- Department of Nutrition and Food Hygiene, School of Public HealthJilin UniversityChangchunChina
| | - Lianzhi Cui
- Department of Clinical LaboratoryJilin Cancer HospitalChangchunChina
| | - Hui Wang
- Department of Physical and Chemical TestJilin Provincial Center for Disease Control and PreventionChangchunChina
| | - Ya Liu
- Department of Nutrition and Food Hygiene, School of Public HealthJilin UniversityChangchunChina
| | - Weiwei Cui
- Department of Nutrition and Food Hygiene, School of Public HealthJilin UniversityChangchunChina
| |
Collapse
|
128
|
Li B, Jia G, Wen D, Zhao X, Zhang J, Xu Q, Zhao X, Jiang N, Liu Z, Wang Y. Rumen microbiota of indigenous and introduced ruminants and their adaptation to the Qinghai-Tibetan plateau. Front Microbiol 2022; 13:1027138. [PMID: 36299720 PMCID: PMC9589358 DOI: 10.3389/fmicb.2022.1027138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/20/2022] [Indexed: 12/03/2022] Open
Abstract
The grassland in the Qinghai-Tibetan plateau provide habitat for many indigenous and introduced ruminants which perform important ecological functions that impact the whole Qinghai-Tibetan plateau ecosystem. These indigenous Tibetan ruminants have evolved several adaptive traits to withstand the severe environmental conditions, especially cold, low oxygen partial pressure, high altitude, strong UV radiation, and poor forage availability on the alpine rangelands. Despite the challenges to husbandry associated with the need for enhanced adaptation, several domesticated ruminants have also been successfully introduced to the alpine pasture regions to survive in the harsh environment. For ruminants, these challenging conditions affect not only the host, but also their commensal microbiota, especially the diversity and composition of the rumen microbiota; multiple studies have described tripartite interactions among host-environment-rumen microbiota. Thus, there are significant benefits to understanding the role of rumen microbiota in the indigenous and introduced ruminants of the Qinghai-Tibetan plateau, which has co-evolved with the host to ensure the availability of specific metabolic functions required for host survival, health, growth, and development. In this report, we systemically reviewed the dynamics of rumen microbiota in both indigenous and introduced ruminants (including gut microbiota of wild ruminants) as well as their structure, functions, and interactions with changing environmental conditions, especially low food availability, that enable survival at high altitudes. We summarized that three predominant driving factors including increased VFA production, enhanced fiber degradation, and lower methane production as indicators of higher efficiency energy harvest and nutrient utilization by microbiota that can sustain the host during nutrient deficit. These cumulative studies suggested alteration of rumen microbiota structure and functional taxa with genes that encode cellulolytic enzymes to potentially enhance nutrient and energy harvesting in response to low quality and quantity forage and cold environment. Future progress toward understanding ruminant adaptation to high altitudes will require the integration of phenotypic data with multi-omics analyses to identify host-microbiota co-evolutionary adaptations enabling survival on the Qinghai-Tibetan plateau.
Collapse
Affiliation(s)
- Bin Li
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
- Agricultural College, Ningxia University, Yinchuan, China
| | - Gaobin Jia
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
- Colleges of Life Science and Technology, Dalian University, Dalian Economic Technological Development Zone, Dalian, China
| | - Dongxu Wen
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Xiuxin Zhao
- Agricultural College, Ningxia University, Yinchuan, China
| | - Junxing Zhang
- Agricultural College, Ningxia University, Yinchuan, China
| | - Qing Xu
- Institute of Life Sciences and Bio-Engineering, Beijing Jiaotong University, Beijing, China
| | - Xialing Zhao
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Nan Jiang
- Colleges of Life Science and Technology, Dalian University, Dalian Economic Technological Development Zone, Dalian, China
| | - Zhenjiang Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Yachun Wang
- Agricultural College, Ningxia University, Yinchuan, China
- Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture of China, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
129
|
Wang C, Cancino A, Baste J, Marten D, Joshi AA, Nasreen A, Bhushan A. Causative Role of Anoxic Environment in Bacterial Regulation of Human Intestinal Function. Cell Mol Bioeng 2022; 15:493-504. [PMID: 36444344 PMCID: PMC9700550 DOI: 10.1007/s12195-022-00735-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/11/2022] [Indexed: 11/03/2022] Open
Abstract
Introduction Life on Earth depends on oxygen; human tissues require oxygen signaling, whereas many microorganisms, including bacteria, thrive in anoxic environments. Despite these differences, human tissues and bacteria coexist in close proximity to each other such as in the intestine. How oxygen governs intestinal-bacterial interactions remains poorly understood. Methods To address to this gap, we created a dual-oxygen environment in a microfluidic device to study the role of oxygen in regulating the regulation of intestinal enzymes and proteins by gut bacteria. Two-layer microfluidic devices were designed using a fluid transport model and fabricated using soft lithography. An oxygen-sensitive material was integrated to determine the oxygen levels. The intestinal cells were cultured in the upper chamber of the device. The cells were differentiated, upon which bacterial strains, a facultative anaerobe, Escherichia coli Nissle 1917, and an obligate anaerobe, Bifidobacterium Adolescentis, were cultured with the intestinal cells. Results The microfluidic device successfully established a dual-oxygen environment. Of particular importance in our findings was that both strains significantly upregulated mucin proteins and modulated several intestinal transporters and transcription factors but only under the anoxic-oxic oxygen gradient, thus providing evidence of the role of oxygen on bacterial-epithelial signaling. Conclusions Our work that integrates cell and molecular biology with bioengineering presents a novel strategy to engineer an accessible experimental system to provide tailored oxygenated environments. The work could provide new avenues to study intestine-microbiome signaling and intestinal tissue engineering, as well as a novel perspective on the indirect effects of gut bacteria on tissues including tumors. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-022-00735-x.
Collapse
Affiliation(s)
- Chengyao Wang
- Department of Biomedical Engineering, Illinois Institute of Technology, 3255 S. Dearborn Street, Wishnick Hall, Chicago, IL 60616 USA
| | - Andrea Cancino
- Department of Biomedical Engineering, Illinois Institute of Technology, 3255 S. Dearborn Street, Wishnick Hall, Chicago, IL 60616 USA
| | - Jasmine Baste
- Department of Biomedical Engineering, Illinois Institute of Technology, 3255 S. Dearborn Street, Wishnick Hall, Chicago, IL 60616 USA
| | - Daniel Marten
- Department of Biomedical Engineering, Illinois Institute of Technology, 3255 S. Dearborn Street, Wishnick Hall, Chicago, IL 60616 USA
| | - Advait Anil Joshi
- Department of Biomedical Engineering, Illinois Institute of Technology, 3255 S. Dearborn Street, Wishnick Hall, Chicago, IL 60616 USA
| | - Amreen Nasreen
- Department of Biomedical Engineering, Illinois Institute of Technology, 3255 S. Dearborn Street, Wishnick Hall, Chicago, IL 60616 USA
| | - Abhinav Bhushan
- Department of Biomedical Engineering, Illinois Institute of Technology, 3255 S. Dearborn Street, Wishnick Hall, Chicago, IL 60616 USA
| |
Collapse
|
130
|
Akkermansia muciniphila: paradigm for next-generation beneficial microorganisms. Nat Rev Gastroenterol Hepatol 2022; 19:625-637. [PMID: 35641786 DOI: 10.1038/s41575-022-00631-9] [Citation(s) in RCA: 522] [Impact Index Per Article: 174.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2022] [Indexed: 12/15/2022]
Abstract
Ever since Akkermansia muciniphila was discovered and characterized two decades ago, numerous studies have shown that the lack or decreased abundance of this commensal bacterium was linked with multiple diseases (such as obesity, diabetes, liver steatosis, inflammation and response to cancer immunotherapies). Although primarily based on simple associations, there are nowadays an increasing number of studies moving from correlations to causality. The causal evidence derived from a variety of animal models performed in different laboratories and recently was also recapitulated in a human proof-of-concept trial. In this Review, we cover the history of the discovery of A. muciniphila and summarize the numerous findings and main mechanisms of action by which this intestinal symbiont improves health. A comparison of this microorganism with other next-generation beneficial microorganisms that are being developed is also made.
Collapse
|
131
|
Yuan Z, Pang Z, Fallah N, Zhou Y, Dong F, Lin W, Hu C. Silicon fertilizer mediated structural variation and niche differentiation in the rhizosphere and endosphere bacterial microbiome and metabolites of sugarcane. Front Microbiol 2022; 13:1009505. [PMID: 36246262 PMCID: PMC9560586 DOI: 10.3389/fmicb.2022.1009505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/08/2022] [Indexed: 12/01/2022] Open
Abstract
The microbiomes of plant are potential determinants of plant growth, productivity, and health. They provide plants with a plethora of functional capacities, namely, phytopathogens suppression, access to low-abundance nutrients, and resistance to environmental stressors. However, a comprehensive insight into the structural compositions of the bacterial abundance, diversity, richness, and function colonizing various microenvironments of plants, and specifically their association with bioactive compounds and soil edaphic factors under silicon (Si) amendment remains largely inconclusive. Here, high-throughput sequencing technology and nontargeted metabolite profiling method were adopted to test the hypotheses regarding microbiome niche abundance, diversity, richness, function, and their association with bioactive compounds and soil edaphic factors within different ecological niches (leaf, stem, root, rhizosphere, and bulk soils) under Si amendment during cane growth were we addressed. Our results demonstrated that Si correspondingly increased sugarcane theoretical production and yield, and remarkably enhanced soil nutrient status, especially Si, AP, and AK. It was also observed that bacterial diversity demonstrated tissue-dependent distribution patterns, with the bulk soil, rhizosphere soil, and root endosphere revealing the highest amount of bacterial diversity compared with the stem and leaf tissues. Moreover, Si exhibited the advantage of considerably promoting bacterial abundance in the various plant compartments. Co-occurrence interactions demonstrated that Si application has the potential to increase bacterial diversity maintenance, coexistence, and plant–soil systems bacteria connections, thereby increasing the functional diversity in the various plant tissues, which, in turn, could trigger positive growth effects in plants. Network analysis further revealed that metabolite profiles exhibited a strong association with bacterial community structures. It was also revealed that Si content had a considerable positive association with bacterial structures. Our findings suggest that the dynamic changes in microbe’s community composition in different plant and soil compartments were compartment-specific. Our study provides comprehensive empirical evidence of the significance of Si in agriculture and illuminated on differential metabolite profiles and soil microbe’s relationship.
Collapse
Affiliation(s)
- Zhaonian Yuan
- Key Laboratory of Sugarcane Biology and Genetic Breeding, Ministry of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Agricultural, Fujian Agriculture and Forestry University, Fuzhou, China
- Province and Ministry Co-sponsored Collaborative Innovation Center of Sugar Industry, Nanning, China
- *Correspondence: Zhaonian Yuan,
| | - Ziqin Pang
- Key Laboratory of Sugarcane Biology and Genetic Breeding, Ministry of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Agricultural, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Nyumah Fallah
- Key Laboratory of Sugarcane Biology and Genetic Breeding, Ministry of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Agricultural, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yongmei Zhou
- College of Agricultural, Fujian Agriculture and Forestry University, Fuzhou, China
- Center for Genomics and Biotechnology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Fei Dong
- Center for Genomics and Biotechnology, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Wenxiong Lin
- College of Agricultural, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Chaohua Hu
- Key Laboratory of Sugarcane Biology and Genetic Breeding, Ministry of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Agricultural, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
132
|
Ling N, Zhang X, Forsythe S, Zhang D, Shen Y, Zhang J, Ding Y, Wang J, Wu Q, Ye Y. Bacteroides fragilis ameliorates Cronobacter malonaticus lipopolysaccharide-induced pathological injury through modulation of the intestinal microbiota. Front Immunol 2022; 13:931871. [PMID: 36211338 PMCID: PMC9536467 DOI: 10.3389/fimmu.2022.931871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/08/2022] [Indexed: 11/23/2022] Open
Abstract
Cronobacter has attracted considerable attention due to its association with meningitis and necrotizing enterocolitis (NEC) in newborns. Generally, lipopolysaccharide (LPS) facilitates bacterial translocation along with inflammatory responses as an endotoxin; however, the pathogenicity of Cronobacter LPS and the strategies to alleviate the toxicity were largely unknown. In this study, inflammatory responses were stimulated by intraperitoneal injection of Cronobacter malonaticus LPS into Sprague–Dawley young rats. Simultaneously, Bacteroides fragilis NCTC9343 were continuously fed through gavage for 5 days before or after injection of C. malonaticus LPS to evaluate the intervention effect of B. fragilis. We first checked the morphological changes of the ileum and colon and the intestinal microbiota and then detected the generation of inflammatory factors, including tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and interleukin-10 (IL-10) and the expression of Toll-like receptor 4 (TLR4), occludin, claudin-4, and iNOs. The results indicated that C. malonaticus LPS exacerbated intestinal infection by altering gut microbe profile, tight junction protein expression, and releasing inflammatory factors in a time- and dose-dependent manner. Intriguingly, treatment with B. fragilis obviously diminished the pathological injuries and expression of TLR4 caused by C. malonaticus LPS while increasing gut microbes like Prevotella-9. We note that Shigella, Peptoclostridium, and Sutterella might be positively related to C. malonaticus LPS infection, but Prevotella-9 was negatively correlated. The results suggested that the intestinal microbiota is an important target for the prevention and treatment of pathogenic injuries induced by C. malonaticus LPS.
Collapse
Affiliation(s)
- Na Ling
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Xiyan Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | | | - Danfeng Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yizhong Shen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Yu Ding
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Juan Wang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- *Correspondence: Yingwang Ye, ; Qingping Wu,
| | - Yingwang Ye
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- *Correspondence: Yingwang Ye, ; Qingping Wu,
| |
Collapse
|
133
|
Akkermansia muciniphila Suppresses High-Fat Diet-Induced Obesity and Related Metabolic Disorders in Beagles. Molecules 2022; 27:molecules27186074. [PMID: 36144806 PMCID: PMC9505756 DOI: 10.3390/molecules27186074] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Obesity is one of the prevalent chronic diseases in human and companion animals usually associated with several metabolic disorders. The gut commensal bacterium Akkermansia muciniphila (A. muciniphila) is known for its therapeutic effects on metabolic disorders and inflammations. Here, we isolated the A. muciniphila AKK2 strain from the feces of interferon-inducible protein 204−/− (IFI204−/−) mice and further evaluated its anti-obesity effects on high-fat diet (HFD)-fed C57BL/6J mice and beagles. The results showed that it effectively controlled weight gain. Microbiome analysis using 16S rRNA gene sequencing revealed that HFD alters gut microbiota composition and A. muciniphila AKK2 increases the Firmicutes/Bacteroidetes (F/B) ratio in beagles. Furthermore, we prepared microcapsules containing A. muciniphila AKK2, and tolerance tests showed the encapsulation maintained high viability and stability in an aerobic environment and simulated the secretion of gastrointestinal fluids. Overall, this study widens the spectrum of A. muciniphila applications to prevent obesity.
Collapse
|
134
|
Chang D, Hu X, Ma Z. Pea-Resistant Starch with Different Multi-scale Structural Features Attenuates the Obesity-Related Physiological Changes in High-Fat Diet Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:11377-11390. [PMID: 36026466 DOI: 10.1021/acs.jafc.2c03289] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The present study compared the modulatory effects of different resistant starches (RSs) isolated from native (NP-RS), acid-hydrolyzed (AHP-RS), and pullulanase debranched (PDP-RS) pea starches on the corresponding in vivo metabolic responses in high fat (HF)-diet-induced obese mice. The biochemical studies on serum lipid profile and antioxidant enzyme activities were supported by histological and gene expression analyses, which suggested a potential therapeutic role for RS in regulating obesity, possibly through the production of short-chain fatty acids and the proliferation of some beneficial colonic bacteria, including Allobaculum, Bifidobacterium, Odoribacter, Clostridium, and Prevotella. Particularly, a more pronounced effect of AHP-RS with a higher proportion of the crystalline region and a more ordered double-helical alignment on improving the hyperlipidemic symptoms in obese mice induced by a HF diet was observed. Our analysis revealed that the RS3 samples seemed to be more effective than RS2 in terms of attenuating obesity in mice that were fed a HF diet.
Collapse
Affiliation(s)
- Danni Chang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, Shaanxi, China
| | - Xinzhong Hu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, Shaanxi, China
| | - Zhen Ma
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, Shaanxi, China
| |
Collapse
|
135
|
Rubini E, Schenkelaars N, Rousian M, Sinclair KD, Wekema L, Faas MM, Steegers-Theunissen RPM, Schoenmakers S. Maternal obesity during pregnancy leads to derangements in one-carbon metabolism and the gut microbiota: implications for fetal development and offspring wellbeing. Am J Obstet Gynecol 2022; 227:392-400. [PMID: 35452650 DOI: 10.1016/j.ajog.2022.04.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/04/2022] [Accepted: 04/07/2022] [Indexed: 11/01/2022]
Abstract
A healthy diet before and during pregnancy is beneficial in acquiring essential B vitamins involved in 1-carbon metabolism, and in maintaining a healthy gut microbiota. Each play important roles in fetal development, immune-system remodeling, and pregnancy-nutrient acquisition. Evidence shows that there is a reciprocal interaction between the one-carbon metabolism and the gut microbiota given that dietary intake of B vitamins has been shown to influence the composition of the gut microbiota, and certain gut bacteria also synthesize B vitamins. This reciprocal interaction contributes to the individual's overall availability of B vitamins and, therefore, should be maintained in a healthy state during pregnancy. There is an emerging consensus that obese pregnant women often have derangements in 1-carbon metabolism and gut dysbiosis owing to high intake of nutritiously poor foods and a chronic systemic inflammatory state. For example, low folate and vitamin B12 in obese women coincide with the decreased presence of B vitamin-producing bacteria and increased presence of inflammatory-associated bacteria from approximately mid-pregnancy. These alterations are risk factors for adverse pregnancy outcomes, impaired fetal development, and disruption of fetal growth and microbiota formation, which may lead to potential long-term offspring metabolic and neurologic disorders. Therefore, preconceptional and pregnant obese women may benefit from dietary and lifestyle counseling to improve their dietary nutrient intake, and from monitoring their B vitamin levels and gut microbiome by blood tests and microbiota stool samples. In addition, there is evidence that some probiotic bacteria have folate biosynthetic capacity and could be used to treat gut dysbiosis. Thus, their use as an intervention strategy for obese women holds potential and should be further investigated. Currently, there are many knowledge gaps concerning the relationship between one-carbon metabolism and the gut microbiota, and future research should focus on intervention strategies to counteract B vitamin deficiencies and gut dysbiosis in obese pregnant women, commencing with the use of probiotic and prebiotic supplements.
Collapse
Affiliation(s)
- Eleonora Rubini
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nicole Schenkelaars
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Melek Rousian
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Kevin D Sinclair
- School of Biosciences, University of Nottingham, Nottingham, United Kingdom
| | - Lieske Wekema
- Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marijke M Faas
- Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Sam Schoenmakers
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
136
|
Noureldein M, Nawfal R, Bitar S, Maxwell SS, Khurana I, Kassouf HK, Khuri FR, El-Osta A, Eid AA. Intestinal microbiota regulates diabetes and cancer progression by IL-1β and NOX4 dependent signaling cascades. Cell Mol Life Sci 2022; 79:502. [PMID: 36040503 PMCID: PMC11802975 DOI: 10.1007/s00018-022-04485-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/21/2022] [Accepted: 07/12/2022] [Indexed: 11/03/2022]
Abstract
Diabetes changes the host microbiota, a condition known as dysbiosis. Dysbiosis is an important factor for the pathogenesis of diabetes and colorectal cancer (CRC). We aimed at identifying the microbial signature associated with diabetes and CRC; and identifying the signaling mechanism altered by dysbiosis and leading to CRC progression in diabetes. MKR mice that can spontaneously develop type 2 diabetes were used. For CRC induction, another subset of mice was treated with azoxymethane and dextran sulfate sodium. To identify the role of microbiota, microbiota-depleted mice were inoculated with fecal microbial transplant from diabetic and CRC mice. Further, a mouse group was treated with probiotics. At the end of the treatment, 16S rRNA sequencing was performed to identify microbiota in the fecal samples. Blood was collected, and colons were harvested for molecular, anatomical, and histological analysis. Our results show that diabetes is associated with a microbial signature characterized by reduction of butyrate-forming bacteria. This dysbiosis is associated with gastrointestinal complications reflected by a reduction in colon lengths. These changes are reversed upon treatment with probiotics, which rectified the observed dysbiosis. Inoculation of control mice with diabetic or cancer microbiota resulted in the development of increased number of polyps. Our data also show that inflammatory cytokines (mainly interleukin (IL)-1β) and NADPH oxidase (NOX)4 are over-expressed in the colon tissues of diabetic mice. Collectively our data suggest that diabetes is associated with dysbiosis characterized by lower abundance of butyrate-forming bacteria leading to over-expression of IL-1β and NOX4 leading to gastrointestinal complications and CRC.
Collapse
Affiliation(s)
- Mohamed Noureldein
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut, 1107-2020, Lebanon
- AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Rashad Nawfal
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut, 1107-2020, Lebanon
- AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Sara Bitar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut, 1107-2020, Lebanon
- AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Scott S Maxwell
- Epigenetics in Human Health and Disease, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Ishant Khurana
- Epigenetics in Human Health and Disease, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Hala Kfoury Kassouf
- Department of Pathology, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Fadlo R Khuri
- Department of Internal Medicine, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Assam El-Osta
- Epigenetics in Human Health and Disease, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
- Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Bliss Street, 11-0236, Riad El-Solh, Beirut, 1107-2020, Lebanon.
- AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
137
|
Farnesoid X receptor activation by the novel agonist TC-100 (3α, 7α, 11β-Trihydroxy-6α-ethyl-5β-cholan-24-oic Acid) preserves the intestinal barrier integrity and promotes intestinal microbial reshaping in a mouse model of obstructed bile acid flow. Biomed Pharmacother 2022; 153:113380. [PMID: 36076475 DOI: 10.1016/j.biopha.2022.113380] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/21/2022] [Accepted: 07/06/2022] [Indexed: 12/12/2022] Open
|
138
|
Gut Microbiota Composition in Undernourished Children Associated with Diet and Sociodemographic Factors: A Case–Control Study in Indonesia. Microorganisms 2022; 10:microorganisms10091748. [PMID: 36144350 PMCID: PMC9502830 DOI: 10.3390/microorganisms10091748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/27/2022] [Accepted: 08/28/2022] [Indexed: 12/16/2022] Open
Abstract
Malnutrition, which consists of undernutrition and overnutrition, is associated with gut microbiota composition, diet, and sociodemographic factors. Undernutrition is a nutrient deficiency that that should be identified to prevent other diseases. In this study, we evaluate the gut microbiota composition in undernourished children in association with diet and sociodemographic factors. We observed normal children (n= 20) and undernourished children (n= 20) for ten days in Lombok and Yogyakarta. Diet, sociodemographic factors, and medical records were recorded using food records, screening forms, and standard household questionnaires. Gut microbiota analysis was performed using 16S rRNA gene sequencing targeting the V3–V4 region. The result showed that the undernourished group had lower energy intake. In addition, the undernourished group had lower quality of medical records, parent knowledge, education, and exclusive breastfeeding. Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia were significantly different between normal and undernourished children. Based on LefSe, we determined that Akkermansia is a biomarker for undernourished children. In conclusion, diet and sociodemographic factors affect the gut microbiota composition of undernourished children.
Collapse
|
139
|
Medley BJ, Leclaire L, Thompson N, Mahoney KE, Pluvinage B, Parson MAH, Burke JE, Malaker S, Wakarchuk W, Boraston AB. A previously uncharacterized O-glycopeptidase from Akkermansia muciniphila requires the Tn-antigen for cleavage of the peptide bond. J Biol Chem 2022; 298:102439. [PMID: 36049519 PMCID: PMC9513282 DOI: 10.1016/j.jbc.2022.102439] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 10/27/2022] Open
Abstract
Akkermansia muciniphila is key member of the human gut microbiota, impacting many features of host health. A major characteristic of this bacterium is its interaction with host mucin, which is abundant in the gut environment, and its ability to metabolize mucin as a nutrient source. The machinery deployed by A. muciniphila to enable this interaction appears to be extensive and sophisticated, yet it is incompletely defined. The uncharacterized protein AMUC_1438 is encoded by a gene that was previously shown to be upregulated when the bacterium is grown on mucin. This uncharacterized protein has features suggestive of carbohydrate-recognition and peptidase activity, which led us to hypothesize that it has a role in mucin depolymerization. Here we provide structural and functional support for the assignment of AMUC_1438 as a unique O-glycopeptidase with mucin degrading capacity. O-glycopeptidase enzymes recognize glycans but hydrolyze the peptide backbone and are common in host-adapted microbes that colonize or invade mucus layers. Structural, kinetic, and mutagenic analyses point to a metzincin metalloprotease catalytic motif but specific recognition of a GalNAc residue α-linked to serine or threonine (i.e. the Tn-antigen) within the AMUC_1438 active site. The enzyme catalyzes hydrolysis of the bond immediately N-terminal to the glycosylated residue. Additional modelling analyses suggest the presence of a carbohydrate-binding module that may assist in substrate recognition. We anticipate that these results will be fundamental to a wider understanding of the O-glycopeptidase class of enzymes and how they may contribute to host-adaptation.
Collapse
Affiliation(s)
- Brendon J Medley
- Department of Biochemistry and Microbiology, University of Victoria, PO Box 1700 STN CSC, Victoria, British Columbia, V8W 2Y2, Canada
| | - Leif Leclaire
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Nicole Thompson
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Keira E Mahoney
- Department of Chemistry, Yale University, 350 Edward St., New Haven CT, 06511
| | - Benjamin Pluvinage
- Department of Biochemistry and Microbiology, University of Victoria, PO Box 1700 STN CSC, Victoria, British Columbia, V8W 2Y2, Canada
| | - Matthew A H Parson
- Department of Biochemistry and Microbiology, University of Victoria, PO Box 1700 STN CSC, Victoria, British Columbia, V8W 2Y2, Canada
| | - John E Burke
- Department of Biochemistry and Microbiology, University of Victoria, PO Box 1700 STN CSC, Victoria, British Columbia, V8W 2Y2, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Stacy Malaker
- Department of Chemistry, Yale University, 350 Edward St., New Haven CT, 06511
| | - Warren Wakarchuk
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Alisdair B Boraston
- Department of Biochemistry and Microbiology, University of Victoria, PO Box 1700 STN CSC, Victoria, British Columbia, V8W 2Y2, Canada.
| |
Collapse
|
140
|
Akkermansia muciniphila Reduces Peritonitis and Improves Intestinal Tissue Wound Healing after a Colonic Transmural Defect by a MyD88-Dependent Mechanism. Cells 2022; 11:cells11172666. [PMID: 36078075 PMCID: PMC9454966 DOI: 10.3390/cells11172666] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/17/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Anastomotic leakage is a major complication following colorectal surgery leading to peritonitis, complications, and mortality. Akkermansia muciniphila has shown beneficial effects on the gut barrier function. Whether A. muciniphila reduces peritonitis and mortality during colonic leakage is unknown. Whether A. muciniphila can directly modulate the expression of genes in the colonic mucosa in humans has never been studied. We investigated the effects of a pretreatment (14 days) with live A. muciniphila prior to surgical colonic perforation on peritonitis, mortality, and wound healing. We used mice with an inducible intestinal-epithelial-cell-specific deletion of MyD88 (IEC-MyD88 KO) to investigate the role of the innate immune system in this context. In a proof-of-concept pilot study, healthy humans were exposed to A. muciniphila for 2 h and colonic biopsies taken before and after colonic instillation for transcriptomic analysis. Seven days after colonic perforation, A.-muciniphila-treated mice had significantly lower mortality and severity of peritonitis. This effect was associated with significant improvements of wound histological healing scores, higher production of IL22, but no changes in the mucus layer thickness or genes involved in cell renewal, proliferation, or differentiation. All these effects were abolished in IEC-MyD88 KO mice. Finally, human subjects exposed to A. muciniphila exhibited an increased level of the bacterium at the mucus level 2 h after instillation and significant changes in the expression of different genes involved in the regulation of cell cycling, gene transcription, immunity, and inflammation in their colonic mucosa. A. muciniphila improves wound healing during transmural colonic wall defect through mechanisms possibly involving IL22 signaling and requiring MyD88 in the intestinal cells. In healthy humans, colonic administration of A. muciniphila is well tolerated and changes the expression of genes involved in the immune pathways.
Collapse
|
141
|
Wang B, Liu J, Lei R, Xue B, Li Y, Tian X, Zhang K, Luo B. Cold exposure, gut microbiota, and hypertension: A mechanistic study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 833:155199. [PMID: 35417730 DOI: 10.1016/j.scitotenv.2022.155199] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 06/14/2023]
Abstract
Cold exposure has been recognized as an important risk factor for hypertension, and altered gut microbiota has been reported to be associated with hypertension. We hypothesized that there is a plausible relationship between gut microbiota and cold-induced hypertension (CIH). Therefore, we explored the potential link between the gut microbiota and its metabolites with CIH. Male Sprague-Dawley (SD) rats were randomly divided into the normal temperature group (NT, 20 ± 2 °C) and the cold exposure group (CE, 4 ± 1 °C), and faecal bacteria cross-transplantation was performed after six weeks. We analyzed the gut microbiota of rats using the 16S rDNA sequence and measured the blood pressure of rats and the content of short-chain fatty acids in rat faeces. After six weeks of cold exposure, the CIH rat model was successfully established. The cold exposure reduced the diversity of the gut microbiota, increased the abundance of potentially pathogenic and conditionally pathogenic bacteria (e.g., Quinella, Rothia, and Senegalimassilia genera), and reduced the abundance of beneficial bacteria (e.g., Lactobacillus genus) and butyric acid-producing bacteria (e.g., Lachnospiraceae UCG-008 and Ruminococcaceae UCG-013 genera). Faecal bacteria cross-transplantation altered gut microbiota composition and regulated blood pressure levels. The NT group rats transplanted with CIH rats' faecal bacteria were enriched with certain conditional pathogenic bacteria such as Prevotellaceae UCG-003 genus. The CIH rats transplanted with faecal bacteria from the NT group rats were enriched with beneficial bacteria such as Bacteroides genus. In addition, we found a significant reduction in butyric acid levels in CIH rats, which may be related to the increase in blood pressure. In conclusion, CIH is associated with altered gut microbiota and reduced butyric acid. Our findings provide novel insights for the prevention and treatment of CIH by modulating the gut microbiota through supplementation of beneficial bacteria/butyrate.
Collapse
Affiliation(s)
- Bo Wang
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Jiangtao Liu
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Ruoyi Lei
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Baode Xue
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Yanlin Li
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Xiaoyu Tian
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Kai Zhang
- Department of Environmental Health Sciences, School of Public Health, University at Albany, State University of New York, One University Place, Rensselaer, NY 12144, USA.
| | - Bin Luo
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China.
| |
Collapse
|
142
|
Comparative Genomics and Physiology of Akkermansia muciniphila Isolates from Human Intestine Reveal Specialized Mucosal Adaptation. Microorganisms 2022; 10:microorganisms10081605. [PMID: 36014023 PMCID: PMC9415379 DOI: 10.3390/microorganisms10081605] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/24/2022] [Accepted: 08/07/2022] [Indexed: 01/07/2023] Open
Abstract
Akkermansia muciniphila is a champion of mucin degradation in the human gastrointestinal tract. Here, we report the isolation of six novel strains from healthy human donors and their genomic, proteomic and physiological characterization in comparison to the type-strains A. muciniphila MucT and A. glycaniphila PytT. Complete genome sequencing revealed that, despite their large genomic similarity (>97.6%), the novel isolates clustered into two distinct subspecies of A. muciniphila: Amuc1, which includes the type-strain MucT, and AmucU, a cluster of unassigned strains that have not yet been well characterized. CRISPR analysis showed all strains to be unique and confirmed that single healthy subjects can carry more than one A. muciniphila strain. Mucin degradation pathways were strongly conserved amongst all isolates, illustrating the exemplary niche adaptation of A. muciniphila to the mucin interface. This was confirmed by analysis of the predicted glycoside hydrolase profiles and supported by comparing the proteomes of A. muciniphila strain H2, belonging to the AmucU cluster, to MucT and A. glycaniphila PytT (including 610 and 727 proteins, respectively). While some intrinsic resistance was observed among the A. muciniphila straind, none of these seem to pose strain-specific risks in terms of their antibiotic resistance patterns nor a significant risk for the horizontal transfer of antibiotic resistance determinants, opening the way to apply the type-strain MucT or these new A. muciniphila strains as next generation beneficial microbes.
Collapse
|
143
|
Lv QB, Li S, Zhang Y, Guo R, Wang YC, Peng Y, Zhang XX. A thousand metagenome-assembled genomes of Akkermansia reveal phylogroups and geographical and functional variations in the human gut. Front Cell Infect Microbiol 2022; 12:957439. [PMID: 35982777 PMCID: PMC9378777 DOI: 10.3389/fcimb.2022.957439] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/11/2022] [Indexed: 01/02/2023] Open
Abstract
Akkermansia muciniphila has long been considered to be the only Akkermansia species in the human gut and has been extensively studied. The present study revealed the genomic architecture of Akkermansia in the human gut by analyzing 1,126 near-complete metagenome-assembled genomes, 84 publicly available genomes, and 1 newly sequenced Akkermansia glycaniphila strain from the human gut. We found that 1) the genomes of Akkermansia were clustered into four phylogroups with distinct interspecies similarity and different genomic characteristics and 2) A. glycaniphila GP37, a strain of Akkermansia, was isolated from the human gut, whereas previously, it had only been found in python. Amuc III was present in the Chinese population, and Amuc IV was mainly distributed in Western populations. A large number of gene functions, pathways, and carbohydrate-active enzymes were specifically associated with phylogroups. Our findings based on over a thousand genomes strengthened our previous knowledge and provided new insights into the population structure and ecology of Akkermansia in the human gut.
Collapse
Affiliation(s)
- Qing-Bo Lv
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
- Puensum Genetech Institute, Wuhan, China
| | | | - Yue Zhang
- Puensum Genetech Institute, Wuhan, China
| | | | - Yan-Chun Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yongzheng Peng
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Yongzheng Peng, ; Xiao-Xuan Zhang,
| | - Xiao-Xuan Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
- *Correspondence: Yongzheng Peng, ; Xiao-Xuan Zhang,
| |
Collapse
|
144
|
Esposito S, Testa I, Mariotti Zani E, Cunico D, Torelli L, Grandinetti R, Fainardi V, Pisi G, Principi N. Probiotics Administration in Cystic Fibrosis: What Is the Evidence? Nutrients 2022; 14:3160. [PMID: 35956335 PMCID: PMC9370594 DOI: 10.3390/nu14153160] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 12/25/2022] Open
Abstract
In the last 20 years, gut microbiota in patients with cystic fibrosis (CF) has become an object of interest. It was shown that these patients had gut dysbiosis and this could explain not only the intestinal manifestations of the disease but also part of those involving the respiratory tract. The acquisition of previously unknown information about the importance of some bacteria, i.e., those partially or totally disappeared in the gut of CF patients, in the regulation of the activity and function of the gut and the lung was the base to suggest the use of probiotics in CF patients. The main aim of this paper is to discuss the biological basis for probiotic administration to CF patients and which results could be expected. Literature analysis showed that CF intestinal dysbiosis depends on the same genetic mutations that condition the clinical picture of the diseases and is aggravated by a series of therapeutic interventions, such as dietary modifications, the use of antibiotics, and the administration of antacids. All this translates into a significant worsening of the structure and function of organs, including the lung and intestine, already deeply penalized by the genetic alterations of CF. Probiotics can intervene on dysbiosis, reducing the negative effects derived from it. However, the available data cannot be considered sufficient to indicate that these bacteria are essential elements of CF therapy. Further studies that take into account the still unsolved aspects on how to use probiotics are absolutely necessary.
Collapse
Affiliation(s)
- Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.M.Z.); (D.C.); (L.T.); (R.G.)
| | - Ilaria Testa
- Respiratory Unit, Great Ormond Street Hospital for Children, Foundation Trust, London WC1N 1LE, UK; (I.T.); (V.F.); (G.P.)
| | - Elena Mariotti Zani
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.M.Z.); (D.C.); (L.T.); (R.G.)
| | - Daniela Cunico
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.M.Z.); (D.C.); (L.T.); (R.G.)
| | - Lisa Torelli
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.M.Z.); (D.C.); (L.T.); (R.G.)
| | - Roberto Grandinetti
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.M.Z.); (D.C.); (L.T.); (R.G.)
| | - Valentina Fainardi
- Respiratory Unit, Great Ormond Street Hospital for Children, Foundation Trust, London WC1N 1LE, UK; (I.T.); (V.F.); (G.P.)
| | - Giovanna Pisi
- Respiratory Unit, Great Ormond Street Hospital for Children, Foundation Trust, London WC1N 1LE, UK; (I.T.); (V.F.); (G.P.)
| | | |
Collapse
|
145
|
Wei F, Yang X, Zhang M, Xu C, Hu Y, Liu D. Akkermansia muciniphila Enhances Egg Quality and the Lipid Profile of Egg Yolk by Improving Lipid Metabolism. Front Microbiol 2022; 13:927245. [PMID: 35928144 PMCID: PMC9344071 DOI: 10.3389/fmicb.2022.927245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
Akkermansia muciniphila (A. muciniphila) has shown potential as a probiotic for the prevention and treatment of non-alcoholic fatty liver disease in both humans and mice. However, relatively little is known about the effects of A. muciniphila on lipid metabolism, productivity, and product quality in laying hens. In this study, we explored whether A. muciniphila supplementation could improve lipid metabolism and egg quality in laying hens and sought to identify the underlying mechanism. In the first experiment, 80 Hy-Line Brown laying hens were divided into four groups, one of which was fed a normal diet (control group), while the other three groups were administered a high-energy, low-protein diet to induce fatty liver hemorrhagic syndrome (FLHS). Among the three FLHS groups, one was treated with phosphate-buffered saline, one with live A. muciniphila, and one with pasteurized A. muciniphila. In the second experiment, 140 Hy-Line Brown laying hens were divided into two groups and respectively fed a basal diet supplemented or not with A. muciniphila lyophilized powder. The results showed that, in laying hens with FLHS, treatment with either live or pasteurized A. muciniphila efficiently decreased body weight, abdominal fat deposition, and lipid content in both serum and the liver; downregulated the mRNA expression of lipid synthesis-related genes and upregulated that of lipid transport-related genes in the liver; promoted the growth of short-chain fatty acids (SCFAs)-producing microorganisms and increased the cecal SCFAs content; and improved the yolk lipid profile. Additionally, the supplementation of lyophilized powder of A. muciniphila to aged laying hens reduced abdominal fat deposition and total cholesterol (TC) levels in both serum and the liver, suppressed the mRNA expression of cholesterol synthesis-related genes in the liver, reduced TC content in the yolk, increased eggshell thickness, and reshaped the composition of the gut microbiota. Collectively, our findings demonstrated that A. muciniphila can modulate lipid metabolism, thereby, promoting laying hen health as well as egg quality and nutritive value. Live, pasteurized, and lyophilized A. muciniphila preparations all have the potential for use as additives for improving laying hen production.
Collapse
|
146
|
Basbas C, Aly S, Okello E, Karle BM, Lehenbauer T, Williams D, Ganda E, Wiedmann M, Pereira RV. Effect of Intramammary Dry Cow Antimicrobial Treatment on Fresh Cow's Milk Microbiota in California Commercial Dairies. Antibiotics (Basel) 2022; 11:963. [PMID: 35884217 PMCID: PMC9312063 DOI: 10.3390/antibiotics11070963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022] Open
Abstract
This study used 16S rRNA sequencing to evaluate the effects of dry cow antimicrobial therapy on the udder milk microbiota by comparing the microbial populations in milk at dry-off (DRY) (~60 days before calving) and post-partum (FRESH) (4-11 days after calving) from cows receiving an intramammary antibiotic infusion prior to dry-off (IMT) and cows that did not receive treatment (CTL). Milk was collected from 23 cows from the IMT group and 27 cows from the CTL group. IMT and DRY samples had a greater correlation with the genera Brevibacterium and Amaricoccus, and the family Micrococcaceae, when compared to IMT and FRESH samples. CTL group samples collected at DRY had a greater correlation with the genera Akkermansia and Syntrophus, when compared to FRESH samples; no bacterial taxa were observed to have a significant correlation with FRESH samples in the CTL group. DRY samples collected from the CTL group had a greater correlation with the genus Mogibacterium when compared to IMT and CTL samples. For DRY samples collected from the IMT group, a greater correlation with the genus Alkalibacterium when compared to DRY and CTL samples, was observed. The lack of a correlation for FRESH samples between the CTL and IMT treatment groups indicated that intramammary antimicrobial dry cow therapy had no significant effect on the udder milk microbiota post-partum.
Collapse
Affiliation(s)
- Carl Basbas
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA; (C.B.); (S.A.); (E.O.); (T.L.)
| | - Sharif Aly
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA; (C.B.); (S.A.); (E.O.); (T.L.)
- Veterinary Medicine Teaching and Research Center, University of California, Davis, Tulare, CA 93274, USA;
| | - Emmanuel Okello
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA; (C.B.); (S.A.); (E.O.); (T.L.)
- Veterinary Medicine Teaching and Research Center, University of California, Davis, Tulare, CA 93274, USA;
| | - Betsy M. Karle
- Cooperative Extension, Division of Agriculture and Natural Resources, University of California, Orland, CA 95963, USA;
| | - Terry Lehenbauer
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA; (C.B.); (S.A.); (E.O.); (T.L.)
- Veterinary Medicine Teaching and Research Center, University of California, Davis, Tulare, CA 93274, USA;
| | - Deniece Williams
- Veterinary Medicine Teaching and Research Center, University of California, Davis, Tulare, CA 93274, USA;
| | - Erika Ganda
- Department of Food Science, Cornell University, Ithaca, NY 14850, USA; (E.G.); (M.W.)
- Penn State College of Agricultural Sciences, University Park, PA 16802, USA
| | - Martin Wiedmann
- Department of Food Science, Cornell University, Ithaca, NY 14850, USA; (E.G.); (M.W.)
| | - Richard V. Pereira
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA; (C.B.); (S.A.); (E.O.); (T.L.)
| |
Collapse
|
147
|
Chen T, Jia F, Yu Y, Zhang W, Wang C, Zhu S, Zhang N, Liu X. Potential Role of Quercetin in Polycystic Ovary Syndrome and Its Complications: A Review. Molecules 2022; 27:molecules27144476. [PMID: 35889348 PMCID: PMC9325244 DOI: 10.3390/molecules27144476] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 12/18/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common multisystem disease with reproductive, metabolic and psychological abnormalities. It is characterized by a high prevalence rate in women of childbearing age and highly heterogeneous clinical manifestations, which seriously harm women’s physical and mental health. Quercetin (QUR) is a natural compound of flavonoids found in a variety of foods and medicinal plants. It can intervene with the pathologic process of PCOS from multiple targets and channels and has few adverse reactions. It is mentioned in this review that QUR can improve ovulation disorder, relieve Insulin resistance (IR), reduce androgen, regulate lipid metabolism, regulate gut microbiota and improve vascular endothelial function, which is of great significance in the treatment of PCOS.
Collapse
Affiliation(s)
- Tong Chen
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fan Jia
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yue Yu
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Wufan Zhang
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chaoying Wang
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- Department of Gynecology of Traditional Chinese Medicine, Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Shiqin Zhu
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- Department of Gynecology of Traditional Chinese Medicine, Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Nana Zhang
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- Department of Gynecology of Traditional Chinese Medicine, Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xinmin Liu
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- Correspondence:
| |
Collapse
|
148
|
Gradisteanu Pircalabioru G, Liaw J, Gundogdu O, Corcionivoschi N, Ilie I, Oprea L, Musat M, Chifiriuc MC. Effects of the Lipid Profile, Type 2 Diabetes and Medication on the Metabolic Syndrome—Associated Gut Microbiome. Int J Mol Sci 2022; 23:ijms23147509. [PMID: 35886861 PMCID: PMC9318871 DOI: 10.3390/ijms23147509] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 02/08/2023] Open
Abstract
Metabolic syndrome (MetSyn) is a major health problem affecting approximately 25% of the worldwide population. Since the gut microbiota is highly connected to the host metabolism, several recent studies have emerged to characterize the role of the microbiome in MetSyn development and progression. To this end, our study aimed to identify the microbiome patterns which distinguish MetSyn from type 2 diabetes mellitus (T2DM). We performed 16S rRNA amplicon sequencing on a cohort of 70 individuals among which 40 were MetSyn patients. The microbiome of MetSyn patients was characterised by reduced diversity, loss of butyrate producers (Subdoligranulum, Butyricicoccus, Faecalibacterium prausnitzii) and enrichment in the relative abundance of fungal populations. We also show a link between the gut microbiome and lipid metabolism in MetSyn. Specifically, low-density lipoproteins (LDL) and high-density lipoproteins (HDL) display a positive effect on gut microbial diversity. When interrogating the signature of gut microbiota in a subgroup of patients harbouring both MetSyn and T2DM conditions, we observed a significant increase in taxa such as Bacteroides, Clostridiales, and Erysipelotrichaceae. This preliminary study shows for the first time that T2DM brings unique signatures of gut microbiota in MetSyn patients. We also highlight the impact of metformin treatment on the gut microbiota. Metformin administration was linked to changes in Prevotellaceae, Rickenellaceae, and Clostridiales. Further research focusing on the microbiome-metabolome patterns is needed to clarify the exact association of various gut microbial communities with the progression of T2DM and the occurrence of various complications in MetSyn patients.
Collapse
Affiliation(s)
| | - Janie Liaw
- Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK;
| | - Ozan Gundogdu
- Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK;
- Correspondence: (G.G.P.); (O.G.)
| | - Nicolae Corcionivoschi
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast BT9 5PX, UK;
- Faculty of Bioengineering of Animal Resources, Banat University of Agricultural Sciences and Veterinary Medicine—King Michael I of Romania, 300645 Timisoara, Romania
| | | | - Luciana Oprea
- National Institute of Endocrinology C.I. Parhon, 011863 Bucharest, Romania; (L.O.); (M.M.)
| | - Madalina Musat
- National Institute of Endocrinology C.I. Parhon, 011863 Bucharest, Romania; (L.O.); (M.M.)
- Department of Endocrinology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mariana-Carmen Chifiriuc
- Research Institute of University of Bucharest (ICUB), 300645 Bucharest, Romania;
- Romanian Academy, 010071 Bucharest, Romania
| |
Collapse
|
149
|
Sugary vs salty food industry leftovers in postweaning piglets: effects on gut microbiota and intestinal volatile fatty acid production. Animal 2022; 16:100584. [DOI: 10.1016/j.animal.2022.100584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 11/22/2022] Open
|
150
|
Kumar R, Kane H, Wang Q, Hibberd A, Jensen HM, Kim HS, Bak SY, Auzanneau I, Bry S, Christensen N, Friedman A, Rasinkangas P, Ouwehand AC, Forssten SD, Hasselwander O. Identification and Characterization of a Novel Species of Genus Akkermansia with Metabolic Health Effects in a Diet-Induced Obesity Mouse Model. Cells 2022; 11:cells11132084. [PMID: 35805168 PMCID: PMC9265676 DOI: 10.3390/cells11132084] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022] Open
Abstract
Akkermansia muciniphila is a well-known bacterium with the ability to degrade mucin. This metabolic capability is believed to play an important role in the colonization of this bacterium in the gut. In this study, we report the identification and characterization of a novel Akkermansia sp. DSM 33459 isolated from human feces of a healthy donor. Phylogenetic analysis based on the genome-wide average nucleotide identity indicated that the Akkermansia sp. DSM 33459 has only 87.5% similarity with the type strain A. muciniphila ATCC BAA-835. Akkermansia sp. DSM 33459 showed significant differences in its fatty acid profile and carbon utilization as compared to the type strain. The Akkermansia sp. DSM 33459 strain was tested in a preclinical obesity model to determine its effect on metabolic markers. Akkermansia sp. DSM 33459 showed significant improvement in body weight, total fat weight, and resistin and insulin levels. Interestingly, these effects were more pronounced with the live form as compared to a pasteurized form of the strain. The strain showed production of agmatine, suggesting a potential novel mechanism for supporting metabolic and cognitive health. Based on its phenotypic features and phylogenetic position, it is proposed that this isolate represents a novel species in the genus Akkermansia and a promising therapeutic candidate for the management of metabolic diseases.
Collapse
Affiliation(s)
- Ritesh Kumar
- Health & Biosciences, International Flavors & Fragrances, Inc. (IFF), Wilmington, DE 19803, USA; (H.K.); (Q.W.); (H.-S.K.); (A.F.)
- Correspondence: ; Tel.: +1-302-379-4738
| | - Helene Kane
- Health & Biosciences, International Flavors & Fragrances, Inc. (IFF), Wilmington, DE 19803, USA; (H.K.); (Q.W.); (H.-S.K.); (A.F.)
| | - Qiong Wang
- Health & Biosciences, International Flavors & Fragrances, Inc. (IFF), Wilmington, DE 19803, USA; (H.K.); (Q.W.); (H.-S.K.); (A.F.)
| | | | - Henrik Max Jensen
- Health & Biosciences, IFF, 8220 Brabrand, Denmark; (H.M.J.); (S.Y.B.); (N.C.)
| | - Hye-Sook Kim
- Health & Biosciences, International Flavors & Fragrances, Inc. (IFF), Wilmington, DE 19803, USA; (H.K.); (Q.W.); (H.-S.K.); (A.F.)
| | - Steffen Yde Bak
- Health & Biosciences, IFF, 8220 Brabrand, Denmark; (H.M.J.); (S.Y.B.); (N.C.)
| | | | - Stéphanie Bry
- Health & Biosciences, IFF, 86270 Dange, France; (I.A.); (S.B.)
| | - Niels Christensen
- Health & Biosciences, IFF, 8220 Brabrand, Denmark; (H.M.J.); (S.Y.B.); (N.C.)
| | - Andrew Friedman
- Health & Biosciences, International Flavors & Fragrances, Inc. (IFF), Wilmington, DE 19803, USA; (H.K.); (Q.W.); (H.-S.K.); (A.F.)
| | - Pia Rasinkangas
- Health & Biosciences, IFF, 02460 Kantvik, Finland; (P.R.); (A.C.O.); (S.D.F.)
| | - Arthur C. Ouwehand
- Health & Biosciences, IFF, 02460 Kantvik, Finland; (P.R.); (A.C.O.); (S.D.F.)
| | - Sofia D. Forssten
- Health & Biosciences, IFF, 02460 Kantvik, Finland; (P.R.); (A.C.O.); (S.D.F.)
| | | |
Collapse
|